Você está na página 1de 41

Pathophysiology and Tre a t m e n t o f S e p t i c Shock in Neonates

James L. Wynn,
KEYWORDS  Neonate  Sepsis  Shock  Treatment  Pathophysiology
MD
a,

*, Hector R. Wong,

MD

Sepsis or serious infection within the first 4 weeks of life kills more than 1 million newborns globally every year.1 The attack rate for neonatal sepsis is variable (from <1% to >35% of live births) based on gestational age and time of onset (early [<72 hours after birth] or late [>72 hours after birth]).25 Neonates with sepsis may present in or progress to septic shock, exemplified initially by cardiovascular dysfunction requiring fluid resuscitation or inotropic support.6 If the progression of infection cannot be stopped, end-organ damage and death become much more likely. Although the true incidence is not known, a recent retrospective cohort study of 3800 neonates admitted to the neonatal intensive care unit (NICU) in a 6-year period reported septic shock in 1.3% with an associated mortality peaking at 71% for extremely low birth weight (ELBW) neonates less than 1000 g.7 There are few published data regarding the pathophysiology of septic shock in neonates. Previous clinical investigations into neonatal sepsis and shock have largely focused on diagnostic markers. Descriptions of septic shock are predominantly case reports on very small numbers, mixed populations with severe respiratory distress syndrome (RDS) and sepsis, or pediatric studies that included neonates who were not evaluated as a separate group.824
DEFINITIONS OF THE SEPSIS CONTINUUM

In 2005, definitions for pediatric infection, systemic inflammatory response syndrome (SIRS), sepsis, severe sepsis, septic shock, and organ dysfunction were suggested that included term neonates (07 days), newborns (1 week to 1 month) and infants (1 month to 1 year) (Tables 1 and 2).25 Working definitions for the sepsis continuum specific for preterm neonates are needed to provide a uniform basis for clinicians and researchers to study and diagnose severe sepsis in this particularly vulnerable

Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Duke University, 2424 Hock Plaza, Suite 504, DUMC Box 2739, Durham, NC 27710, USA b Division of Critical Care Medicine, Cincinnati Childrens Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA * Corresponding author. E-mail address: james.wynn@duke.edu Clin Perinatol 37 (2010) 439479 doi:10.1016/j.clp.2010.04.002 perinatology.theclinics.com 0095-5108/10/$ see front matter 2010 Elsevier Inc. All rights reserved.

440

Wynn & Wong

Table 1 Definition of systemic inflammatory response syndrome (SIRS), infection, sepsis, severe sepsis, and septic shock Consensus Definitions SIRS The presence of at least 2 of the following 4 criteria, 1 of which must be abnormal temperature or leukocyte count:  Corea temperature of >38.5 C or <36 C  Tachycardia, defined as a mean heart rate >2SD more than normal for age in the absence of external stimulus, chronic drugs, or painful stimuli; or otherwise unexplained persistent increase in a 0.5- to 4-h time period OR for children <1 y old: bradycardia, defined as a mean heart rate <10th percentile for age in the absence of external vagal stimulus, b-blocker drugs, or congenital heart disease; or otherwise unexplained persistent depression in a 0.5-h time period  Mean respiratory rate >2SD more than normal for age or mechanical ventilation for an acute process not related to underlying neuromuscular disease or the receipt of general anesthesia  Leukocyte count increased or decreased for age (not secondary to chemotherapy-induced leukopenia) or >10% immature neutrophils Infection A suspected or proven (by positive culture, tissue stain, or polymerase chain reaction test) infection caused by any pathogen OR a clinical syndrome associated with a high probability of infection. Evidence of infection includes positive findings on clinical examination, imaging, or laboratory tests (eg, white blood cells in a normally sterile body fluid, perforated viscus, chest radiograph consistent with pneumonia, petechial or purpuric rash, or purpura fulminans) Suggested Modifications for Premature Infants SIRS The presence of at least 2 of the following 4 criteria, 1 of which must be abnormal temperature or leukocyte count:  Core temperature of >38.0 Cb or <36 C  Tachycardia, defined as a mean heart rate >2SD more than normal for age in the absence of external stimulus, chronic drugs, or painful stimuli; or otherwise unexplained persistent increase in a 0.5- to 4-h time period OR bradycardia, defined as a mean heart rate <10th percentile for age in the absence of b-blocker drugs or congenital heart diseasec; or otherwise unexplained persistent bradycardiad  Mean respiratory rate >2SD more than normal for age or mechanical ventilation for an acute process not related to underlying neuromuscular disease or the receipt of general anesthesia  Leukocyte count increased or decreased for age or >20% immature to total neutrophil ratioe or C-reactive protein >10 mg/dL No change suggested

Sepsis SIRS in the presence of or as a result of suspected or proven infection Severe sepsis Sepsis plus 1 of the following: cardiovascular organ dysfunction OR ARDS OR 2 or more other organ dysfunctions Septic shock Sepsis and cardiovascular organ dysfunction
a b

No change suggested No change suggested

No change suggested

Core temperature must be measured by rectal, bladder, oral, or central catheter probe. Neonatal fever is considered greater than 38 C. c External vagal stimulus use is uncommon in preterm infants. d Infrequent self-resolving bradycardic episodes can be common in premature neonates in the absence of sepsis. e More commonly accepted ratio is greater than 20% immature to total ratio and chemotherapy-induced leukopenia is uncommon in premature infants. From Goldstein B, Giroir B, Randolph A. International pediatric sepsis consensus conference: definitions for sepsis and organ dysfunction in pediatrics. Pediatr Crit Care Med 2005;6(1):28; with permission.

Treatment of Septic Shock in Neonates


441

442

Wynn & Wong

Table 2 Definitions of organ dysfunction Consensus Definitions of Organ Dysfunction25 Cardiovascular dysfunction Despite administration of isotonic intravenous fluid bolus >40 mL/kg in 1 h  Decrease in BP (hypotension) <5th percentile for age or systolic BP >2SD less than normal for age OR  Need for vasoactive drug to maintain BP in normal range (dopamine >5 mg/kg/min or dobutamine, epinephrine, or norepinephrine at any dose) OR  Two of the following: Unexplained metabolic acidosis: base deficit >5.0 mEq/L Increased arterial lactate >2 times upper limit of normal Oliguria: urine output <0.5 mL/kg/h Prolonged capillary refill >5 s Core to peripheral temperature gap >3 C Pulmonarya  PaO2/FIO2 <300 in absence of cyanotic heart disease or preexisting lung disease OR  PaCO2 >65 torr or 20 mm Hg more than baseline PaCO2 OR  Proven needb for >50% FIO2 to maintain saturation >92% OR  Need for nonelective invasive or noninvasive mechanical ventilationc Suggested Modifications for Premature Infants Cardiovascular dysfunction Despite administration of isotonic intravenous fluid bolus >40 mL/kg in 1 h (>10 ml/kg in infants <32 weeks)d  Decrease in BP (hypotension) <5th percentile for age or systolic BP >2SD less than normal for age or MAP <30 mm Hg with poor capillary refill time (>4 s)e OR  Need for vasoactive drug to maintain BP in normal range (dopamine >5 mg/kg/min or dobutamine, or epinephrine at any dose)f OR  Two of the following: Unexplained metabolic acidosis: base deficit >5.0 mEq/L Increased arterial lactate >2 times upper limit of normal Oliguria: urine output <0.5 mL/kg/h Prolonged capillary refill >4 sg Simultaneous measurement of core and peripheral temperature not common in premature neonates Pulmonary  Excessive oxygen should be limited to avoid complications including retinopathy of prematurity  PaCO2 >65 torr or 20 mm Hg more than baseline PaCO2 OR  Proven need for >50% FIO2 to maintain saturation >92% (88% for <32 weeks) OR  Need for nonelective invasive or noninvasive mechanical ventilation

Neurologic  Glasgow Coma Score >11 OR  Acute change in mental status with a decrease in Glasgow Coma Score >3 points from abnormal baseline Hematologic  Platelet count <80,000/mm3 or a decline of 50% in platelet count from highest value recorded in the past 3 days (for chronic hematology/oncology patients) OR  International normalized ratio >2 Renal  Serum creatinine >2 times upper limit of normal for age or 2-fold increase in baseline creatinine Hepatic  Total bilirubin >4 mg/dL (not applicable for newborn) OR  ALT 2 times upper limit of normal for age

Neurologic  Acute change in mental statush

Hematologic  Platelet count <80,000/mm3 or a decline of 50% in platelet count from highest value recorded in the past 3 daysi OR  International normalized ratio >2 Renal  Serum creatinine >2 times upper limit of normal for age or 2-fold increase in baseline creatinine Hepatic  ALT 2 times upper limit of normal for agej or 50% increase over patients baselinek

Treatment of Septic Shock in Neonates

Abbreviations: ALT, alanine transaminase; BP, blood pressure. a ARDS must include a PaO2/FIO2 ratio %200 mm Hg, bilateral infiltrates, acute onset, and no evidence of left heart failure. Acute lung injury is defined identically except the PaO2/FIO2 ratio must be R300 mm Hg. b Proven need assumes oxygen requirement was tested by decreasing flow with subsequent increase in flow if required. c In postoperative patients, this requirement can be met if the patient has developed an acute inflammatory or infectious process in the lungs that prevents them from being extubated. d Rapid large volume expansion can be associated with intraventricular hemorrhage. e 30 mm Hg suggested as minimum MAP. f Norepinephrine not commonly used in premature neonates. g Greater than 4 s may reflect a low systemic blood flow.264 h Glasgow Coma Score not applicable to term or preterm neonates. i Neonates not frequently chronic hematology-oncology patients. j Indirect hyperbilirubinemia is common in newborns. k Transaminases are commonly increased in preterm neonates on long-term intravenous hyperalimentation. From Goldstein B, Giroir B, Randolph A. International pediatric sepsis consensus conference: definitions for sepsis and organ dysfunction in pediatrics. Pediatr Crit Care Med 2005;6(1):28; with permission.

443

444

Wynn & Wong

population. The authors have proposed modifications to the consensus definitions to incorporate preterm infants that are also presented in Tables 1 and 2. Why have definitions of sepsis and septic shock not been established for preterm neonates? These patients present diagnostic challenges that are clouded by immaturity of organ systems and transitional physiology. For example, normal blood pressure values for gestational and postnatal age have not been established, particularly in the very low birth weight neonate (VLBW, <1500 g), largely because blood pressure alone cannot identify abnormal cardiac output, organ perfusion, and oxygen delivery.26 In the absence of normative values, it is nearly impossible to establish parameters that are associated with poor outcome. Perhaps the most obvious limitation is the differences in monitoring capabilities between the preterm neonate and older, physically larger patients. For example, pulmonary artery catheterization may be used in children or adults to monitor the course of septic shock, but this is not feasible in small neonates. For these reasons, the hemodynamic response to septic shock and optimum clinical interventions in preterm neonates are not well understood.
RISK FACTORS FOR DEVELOPMENT OF NEONATAL SEPTIC SHOCK

Risk factors for a neonate developing sepsis have been well described. Although risk factors for septic shock overlap those for sepsis, specific antenatal and postnatal risks for the development of neonatal septic shock have not been described in detail. Maternal factors contributing to the risk of neonatal sepsis are shown in Box 1 and include prematurity, low birth weight, rectovaginal colonization with group B streptococcus (GBS), prolonged rupture of membranes, maternal intrapartum fever, and chorioamnionitis.2,3,2733 Factors in the postnatal period associated with an increased risk of sepsis or septic shock include male gender, birth weight less than 1000 g, hypogammaglobulinemia, intravenous alimentation, central venous catheters, use of steroids or drugs that decrease gastric acid acidity, and prolonged duration of mechanical ventilation. The development of severe necrotizing enterocolitis (NEC) is also associated with severe sepsis, shock, multi-organ system failure, and death.34,35 Genetic evaluations in children and adults have identified several polymorphisms in cytokines and their receptors as well as other host defense proteins that may either increase or decrease risk for sepsis or poor outcome from sepsis.3641 However, gene polymorphism studies in neonates have not yielded consistent results because of relatively small sample sizes and a general lack of formal prospective validation studies.4256
MICROBIOLOGY OF SEPSIS AND SEPTIC SHOCK IN NEONATES

Several pathogens have been associated with sepsis in the neonatal period. The predominant agents are bacterial, but viruses including herpes simplex and enteroviruses have been associated with fulminant neonatal sepsis with high mortality.5759 In 1 study, gram-negative infection accounted for 38% of cases of septic shock and 62.5% of sepsis mortality.7 These results are similar to those from a previous study that showed gram-negative infection was associated with 69% of cases of fulminant septic shock (death within 48 hours).60 Gram-positive causes of sepsis are dominated by GBS and coagulase-negative staphylococcus (CoNS).3,61 Although lethality and shock from GBS have been well described, mortality associated with CoNS is extremely low3,4 and septic shock is rare.60 Fungi (primarily Candida albicans) may also lead to fulminant neonatal sepsis and predominantly affect ELBW infants.3,62,63 Studies of neonatal sepsis are confounded by the limitations of sensitivity of the current diagnostic gold standard blood culture. Sample volume constraints in

Treatment of Septic Shock in Neonates

445

Box 1 Risk factors for the development of neonatal sepsis and septic shock Maternal factors  Maternal age (>30 years)  Lack of prenatal care  High gravidity  Premature or prolonged (>6 hours) rupture of membrane (PROM)  Meconium-stained amniotic fluid  Foul-smelling amniotic fluid  Premature labor  Chorioamnionitis  GBS rectovaginal colonization  Urinary tract infection  Intrapartum fever  Multiple courses of prenatal steroids or tocolytic agents  Prolonged duration of internal monitoring Delivery room  Prematurity <37 weeks  Low birth weight %2500 g  5-minute Apgar score <5  Resuscitation in delivery room  Male gender Neonatal  Vascular catheterization  Mechanical ventilation (continuous positive airway pressure or endotracheal tube)  Lack of enteral feeding  Gastrointestinal tract pathology  Medications (H2 blockers, proton pump inhibitors; postnatal steroids, cephalosporins)  Neutropenia  Decreased baseline serum IgG concentrations  Hyperalimentation  Prolonged hospital stay  Delay in time to regain birth weight

newborns may undermine the identification of organisms causing shock, particularly in preterm infants.64 For this reason, many studies combine the entities culture-proven sepsis and clinical sepsis (cultures negative but strong clinical suspicion leading to long-term antibiotic treatment). Improved techniques such as molecular diagnostics, see the article by Benitz elsewhere in this issue for further exploration of this topic, may help to delineate which patients with clinical sepsis truly have sepsis versus other causes of clinical deterioration.

446

Wynn & Wong

PATHOPHYSIOLOGY OF SEPSIS AND SHOCK: MOLECULAR AND CELLULAR EVENTS Molecular Signaling: Pattern Recognition Receptors, Pathogen-associated Molecular Patterns, and Damage- or Danger-associated Molecular Patterns

Pathogen recognition by local immune sentinel cells is the first step toward the development of an immune response once local barrier function has been compromised (Fig. 1). Recognition is initiated via the activation of pattern recognition receptors (PRRs)65 including Toll-like receptors (TLRs). There are 10 known TLRs in humans, and each receptor has a specific molecular activation trigger.66,67 TLRs, present on and within multiple cell types, recognize extracellular and intracellular pathogens by their signature microbial products known as pathogen-associated molecular patterns (PAMPs). Lipopolysaccharide (LPS, endotoxin) on gram-negative bacteria is the prototypic PAMP and a key mediator of systemic inflammation, septic shock, and multi-organ failure and death.68 LPS signals primarily through TLR4 in conjunction with the cell surface adaptor proteins CD14 and MD-2.65 Gram-positive bacterial PAMPS such as lipoteichoic acid signal primarily through TLR2, whereas viral PAMPS such as double-stranded RNA signal through TLR3. Microorganisms often stimulate more than 1 TLR simultaneously allowing for initiation of a pathogen-specific host response.67,69 Ligand-receptor binding results in downstream production of cytokines and chemokines as well as activation of other antimicrobial effector mechanisms.66 Intracellular non-TLR PRRs include NOD-like receptors (NLRs) and RIG-like receptors (RLRs). Nucleotide-binding oligomerization domain (an NLR) detects peptidoglycan of gram-positive bacteria in the cytosol, and retinoic acidinducible protein I (RIG-I) detects viral double-stranded RNA and induces type I interferon production.67 Once engaged by pathogens, these PRRs initiate an immune response including the production of proinflammatory cytokines via mitogen-activated protein kinase (MAPK) and the transcription factor nuclear factor kB (NF-kB). To date, RLR and NLR function have not been examined in neonates with sepsis. Because TLRs play an essential role in recognition and response to pathogens, alterations in their expression, structure, signaling pathways, and function can have consequences to host defense. Polymorphisms or mutations in TLRs are associated with increased risk for infection in adults7073 and in children7476 but are less well characterized in neonates. Upregulation of TLR2 and TLR4 mRNA in leukocytes of neonates occurs during gram-positive and gram-negative infection, respectively, across gestational ages.77 Dysregulation or overexpression of TLR4 is involved in the development of necrotizing enterocolitis in experimental animal models,78 demonstrating the importance of TLRs in the initial immune response to pathogens and their role in neonatal sepsis and septic shock. Mutations have been identified in NLRs that are involved in the pathogenesis of neonatal-onset multisystem inflammatory disease (cryopyrin).79 Investigation for mutations in specific domains of NLRs has been performed to identify causes of abnormal inflammatory signaling leading to NEC, but no associations have been identified.80 RLR mutations have been identified but are of unknown clinical significance.81 The role that intracellular PRR play is of particular interest with respect to defense against Listeria monocytogenes, a pathogen particularly virulent in neonates, which can be recognized by NLRs.82 Mutations or decreased expression of costimulatory molecules necessary for TLR activation are also associated with an increased risk for infection. For example, the lipopolysaccharide (LPS, endotoxin) coreceptor CD14 and LPS-binding protein (LBP, which binds intravascular LPS and facilitates its attachment to CD14) are both increased during neonatal sepsis.8385 Genetic variations in these proteins have been associated with increased risk for sepsis in adults.47,49,50 Gene

Treatment of Septic Shock in Neonates

Fig. 1. Activation of sentinel immune cells. Sentinel cells (e.g. monocyte, macrophage) sense pathogens via Pathogen-associated molecular patterns (PAMPs) or damage-/danger-associated molecular patterns (DAMPs) binding to PRRs. PRRs include Toll-like receptors (TLRs), Rig-1-like receptors (RLRs), and NOD-like receptors (NLRs). PAMPs include lipopolysaccharide (LPS), lipotechoic acid (LTA), DNA, and RNA. DAMPs can also be sensed through TLRs and include uric acid (UA), heat shock proteins (Hsp), and HMGB-1. Signaling occurs through a series of second messengers and results in transcription and translation of cytokines and chemokines that amplify the immune response.

447

448

Wynn & Wong

polymorphisms in myeloid differentiation-2 (MD-2), a small protein involved in LPS signaling through TLR4, increase the risk for organ dysfunction and sepsis in adults86 but the significance in neonates is unknown. Polymorphisms in select cytokines (IL-6 and IL-10) or their receptors (IL-4ra53), and constituents of their signaling pathways, may be associated with increased risk of infection,42,43,46,51 although there is not complete agreement on these findings.44,52,54 Polymorphisms in post-TLR activation intracellular signaling molecules including myeloid differentiation factor 88 (MyD88),87 IL-1 receptorassociated kinase 4 (IRAK-4),88 and NF-kB essential modulator (NEMO)89 are associated with invasive bacterial infection in older populations. These genetic factors predisposing to sepsis are likely just the tip of the iceberg because evaluation of intracellular second messenger inflammatory signaling systems is a relatively new and active area of research. In addition to being activated by PAMPs, TLRs can be activated by damage- or danger-associated molecular patterns (DAMPs), such as intracellular proteins or mediators released by dying or damaged cells (see Fig. 1). High-mobility group box-1 (HMGB-1), an important DAMP, is involved in the progression of sepsis to septic shock.68,90 HMGB-1 is produced by macrophages or endothelial cells stimulated with LPS or TNF-a and signals through TLR2, TLR4, and receptor for advanced glycation end products (RAGE).91 Important actions of HMGB-1 include cytokine production, activation of coagulation, and neutrophil recruitment.90,92 HMGB-1 mediates disruption of epithelial junctions within the gut via the induction of reactive nitrogen intermediates (RNI) leading to increased bacterial translocation.93 The role of HMGB-1 and RAGE signaling in septic shock in human neonates has not been well studied, but has been linked to the pathophysiology of NEC in a preclinical model.94 Other DAMPs including heat shock proteins (Hsps) and uric acid may also contribute to the pathophysiology of septic shock. Hsps activate proinflammatory signaling through TLRs, regulate neutrophil function, are immune adjuvants, and are increased in adults and children with sepsis.95 Increased Hsp60 and Hsp70 measured within 24 hours of pediatric intensive care unit admission was associated with pediatric septic shock and there was a strong trend toward a significant association with death.96,97 Hsp production in septic neonates has not been evaluated. Uric acid can increase cytokine production, polymorphonuclear leukocyte (PMN) recruitment, and dendritic cell stimulation,98 and may also serve as an antioxidant.99 Uric acid is reduced in the serum of septic neonates compared with control neonates.100 The importance of DAMPs in neonatal sepsis and shock has yet to be determined.
Cytokines, Chemokines, and Adhesion Molecules

Following PRR stimulation, production of cytokines and chemokines results in amplification of the innate response directed at the invading organisms (see Fig. 1). Increases of proinflammatory cytokines during sepsis and septic shock have been identified including interleukin (IL)-1b, IL-6, IL-8, IL-12, IL-18, interferon gamma (IFN-g), and tumor necrosis factor-alpha (TNF-a).101 Compared with septic adults, septic neonates produce less IL-1b, TNF-a, IFN-g, and IL-12.102107 The decreased cytokine production is due in part to decreased production of important intracellular mediators of TLR signaling including myeloid differentiation factor 88 (MyD88), interferon regulatory factor 5 (IRF5), and p38, which exhibit gestational age-specific diminution.108 In a recent comprehensive study (>140 analytes) of serum from neonates evaluated for late-onset sepsis, IL-18 emerged as a predictive biomarker to differentiate infected from noninfected neonates,109 similar to data from adults with sepsis.110 IL-18 reduces PMN apoptosis,111 potentiates IFN-g production,112 and induces

Treatment of Septic Shock in Neonates

449

production of TNF-a, IL-1b, and IL-8.113 IL-18 primes PMNs for degranulation with production of reactive oxygen intermediates (ROI) on subsequent stimulation.114 Dysregulation of many of these functions linked to IL-18 are seen in sepsis and septic shock. Proinflammatory cytokine production leads to activation of endothelial cells including increased expression of cell adhesion molecules (CAMs) that facilitate leukocyte recruitment and diapedesis (Fig. 2). Upregulation of CAMs (soluble ICAM, VCAM, L-, P-, and E-selectins, and CD11b/CD18) during sepsis facilitates rolling and extravascular migration of leukocytes.115118 Decreased neonatal PMN and monocyte Lselectin and MAC-1 expression impair accumulation at sites of inflammation.119,120 Chemokine gradients produced by endothelial cells and local macrophages are necessary in addition to CAM interactions for effective and specific leukocyte attraction and accumulation. Without adequate leukocyte recruitment, there is increased risk for propagation from a local to a systemic infection. Although poor cellular chemotaxis in the neonate has been observed, it is not likely a result of reduced serum concentrations of chemokines.121 Suboptimal chemotaxis may be related to other mechanisms such as poor complement receptor upregulation following stimulation,122 deficiencies in another downstream signaling process,123 or inhibition by bacterial products.124 A wide variety of chemokines are increased during sepsis including IP-10, CCL5 (RANTES), MCP-1, MIP-1, and IL-8.125 Other chemoattractive molecules are also increased in sepsis including complement proteins C3a and C5a, host defense proteins or peptides such as cathelicidins and defensins, and components of invading bacteria themselves.101,109 The role of chemoattractive substances in the pathogenesis of severe sepsis is highlighted by recent studies showing IL-8 can be used as a stratifying factor for survival in children126 and C5a is implicated in sepsis-associated organ dysfunction in adults.68 Studies of chemokines in neonates with sepsis have shown that IP-10 is a sensitive early marker of infection,125 and decreased levels of CCL5 help predict development of disseminated intravascular coagulation (DIC).127
Antiinflammatory Response

If inflammatory homeostasis is not restored, the consequences can include SIRS, which is associated with multi-organ failure and death (Fig. 3). The careful interplay between anti- and proinflammatory stimuli serves to govern the immune response to allow local pathogen containment but prevent systemic activation leading to excessive inflammatory damage through SIRS.128 Near simultaneous increases in antiinflammatory cytokine production occur during infection, with TGF-b, IL-4, IL-10, IL-11, and IL-13 countering the actions of proinflammatory cytokines (see Fig. 2).101,129,130 These mediators blunt the activation of phagocytic cells, block fever, modify coagulation factor expression, and decrease production of ROI/RNI, NO, and other vasoactive mediators.131135 In addition to the antiinflammatory cytokines, specific soluble cytokines and receptor antagonists produced during sepsis modulate proinflammatory mediator action, including TNFR2 (which regulates the concentration of TNF-a), sIL-6R, sIL2, and IL-1ra. Increases in these inhibitors have been documented in neonatal sepsis with resolution following effective treatment.130,136,137 The role of these regulatory cytokine inhibitors in the immune response to neonatal sepsis and septic shock has been incompletely characterized. Soluble RAGE (sRAGE) competes with cell-bound RAGE for the binding of HMGB-1 and other RAGE ligands,138 reduces the intensity of the inflammatory response, and is increased in adults during sepsis.139 In addition, administration of exogenous sRAGE improved survival and reduced inflammation in infected adult rodents.140

450

Wynn & Wong

Fig. 2. Cellular recruitment and endothelial activation following pathogen detection. Pathogen-stimulated tissue/blood monocytes, dendritic cells (DC), and macrophages release proinflammatory cytokines that activate the surrounding endothelium. Endothelial activation results in upregulation of CAM, production of chemokines and vasoactive substances, activation of complement, and development of a procoagulant state. Recruitment of PMNs occurs along the chemokine gradient surrounding the area of inflammation. Antiinflammatory cytokines counter the actions of proinflammatory cytokines to prevent excessive cellular activation and recruitment that can result in tissue damage and systemic inflammation. Endothelium can be damaged when PMNs release ROI. LTE, leukotriene; NO, nitric oxide; PMN, neutrophil.

Treatment of Septic Shock in Neonates

451

Fig. 3. Pathophysiology of neonatal sepsis and septic shock. AEM, antimicrobial effector mechanisms; CV, cardiovascular; DAMP, danger-/damage-associated molecular patterns; DIC, disseminated intravascular coagulation; PRR, pattern recognition receptors; SIRS, systemic inflammatory response syndrome.

Role of Complement in Host Defense and Sepsis Pathophysiology

Complement is an extraordinarily important component of early innate immunity that facilitates killing of bacteria through opsonization and direct microbicidal activity. Complement components also possess chemotactic or anaphylactic activity that increases leukocyte aggregation and local vascular permeability at the site of invasion. In addition, complement components reciprocally activate several other important processes such as coagulation, proinflammatory cytokine production, and leukocyte activation (see Fig. 3).68 Dysregulation of complement activation may participate in the untoward effects seen in neonates with severe sepsis or septic shock. Neonates, particularly the very premature, exhibit decreased basal levels of complement proteins and function for the alternative and classic pathways.141,142 In addition, complement-mediated opsonization is poor in premature neonates and limited in term neonates.143,144 Complement-mediated activation of leukocytes during sepsis occurs via up-regulated cell surface receptors (CR1 [CD35], CR3 [Mac-1, CD11b/CD18]).145,146 For example, stimulation of CR1 and C5aR, the receptors for C3b and C5a, respectively, facilitate opsonization (CR1-C3b), redistribution of blood flow, increased inflammation, platelet aggregation, and release of ROI (C5a-C5aR).147,148 In addition, activation of the multifunctional CR3 facilitates leukocyte adhesion, phagocytosis, migration and activation, as well as recognition of a broad range of microbial products.149 Upregulation of CR3 on neutrophils following stimulation is blunted in neonates compared with adults and is believed to play a significant role in diminished chemotaxis and transmigration.122 Similar to the effects of TLR stimulation, C5a-mediated local leukocyte activation also results in increased cytokine production with subsequent upregulation of adhesion molecules on

452

Wynn & Wong

vascular endothelium allowing for increased cell recruitment to the site of infection.150 Deficiencies in C5aR found in term neonates compared with adults may limit the ability to respond to C5a and therefore increase the likelihood of infection.151 The expression of C5aR on neutrophils of preterm infants has not been quantified. Complement regulatory proteins modify the effects of complement and prevent potential damage caused by over activation. In particular, CD59 blocks formation of C9 polymerization and target lysis, CD55 destabilizes CR1 and C3 and C5 convertases, and CD35 (CR1) accelerates the deactivation of C3b.152 The role of these regulators in the neonatal response to sepsis and septic shock is presently unknown. Dysregulation of complement activation can lead to a vicious activation cycle that results in excessive cellular stimulation, cytokine production, endothelial cell activation, and local tissue damage. Dysregulation likely contributes to the development of SIRS and shock (see Fig. 3).153 Data in adults link increased C5a levels with multiple facets of sepsis-associated pathology such as the development of DIC via increased tissue factor expression, cardiomyopathy, increased proinflammatory cytokine levels and the development of SIRS, adrenal insufficiency, and neutrophil dysfunction.68 Whether or not C5a or other complement proteins play a role in the development of these phenomena in septic neonates remains to be determined.
Other Host Defense Proteins, Acute Phase Reactants, and Opsonins

In addition to the initial inflammatory response and complement activation following pathogen recognition, the presence of microbes result in increases in other innate proteins that possess valuable immune function.154 These components serve to reduce bacterial load and include collectins (eg, surfactant proteins A and D), lactoferrin, cathelicidins, bacteriocidal permeability increasing protein (BPI), and phospholipase A2.155 Acute phase reactant proteins such as CRP (opsonin), haptoglobin and lactoferrin (reduce available iron/antimicrobial peptide-lactoferricin), serum amyloid A (cellular recruitment), procalcitonin (unknown function), and others increase during sepsis and provide useful ancillary immune functions.101 Neutrophils from term neonates are deficient in BPI, potentially contributing to the increased risk for infection.156 Polymorphisms in BPI increase the risk for gram-negative sepsis in children,157 although the effect of these polymorphisms in neonates is unknown. Sepsis results in an increase in other serum components with opsoninic function including fibronectin and natural antibodies (predominantly IgM) produced by circulating B1 lymphocytes.158160 Despite these increases, neonatal plasma has significantly impaired opsonizing activity compared with adults that increases the likelihood of progression to systemic infection.161
Role of Dysregulated Coagulation in Severe Sepsis

Development of a procoagulant state in the microvasculature surrounding a focal site of infection is a natural host defense mechanism, trapping invading pathogens and preventing further dissemination (see Fig. 2). However, like the inflammatory response, if the procoagulant response to infection escalates unchecked, it can lead to DIC resulting in severe tissue and organ damage (see Fig. 3).162 Neonates with early increased ratios of serum inflammatory to antiinflammatory cytokines during sepsis have an increased risk of developing DIC.127 This finding is consistent with the increased serum levels of IL-659 and high frequency of DIC seen with disseminated HSV infection.163 Initiation of coagulation cascades during infection may begin with activated neutrophils, monocytes, or endothelium, which express increased tissue factor apoprotein.164,165Activation of tissue factor leads to increased clotting proteins including thrombin-antithrombin complex (TAT), plasminogen activator inhibitor

Treatment of Septic Shock in Neonates

453

(PAI), and plasmin-a2-antiplasmin complex.166 There is also a shift toward inactivation of protein S and depletion of anticoagulant proteins including antithrombin III (ATIII) and protein C.167,168 A small study reported that low protein C levels in preterm neonates with sepsis predicted death.169 In DIC, platelets are consumed in microthrombi creating a state of thrombocytopenia; a common finding in infected neonates.170 The longest duration and lowest initial and nadir platelet levels have been noted during neonatal gram-negative and fungal infections,171 and this thrombocytopenia may or may not be associated with DIC. Decreased platelet function in preterm neonates with sepsis further increases the risk for bleeding.172 In ELBW infants, platelets are hyporeactive for the first few days after birth, complicating the ability of the immune system to contain a microbiological threat and increasing the risk for hemorrhage.173
Role of the Neutrophil in Septic Shock

The most important means of early innate cellular defense against bacterial invasion in neonates is the neutrophil or PMN. Neonatal PMNs exhibit quantitative and qualitative deficits compared with adult cells.174,175 A complete discussion of these deficits is presented elsewhere in this issue. Three aspects of PMN function with particular relevance to neonatal severe sepsis and septic shock deserve brief mention: neutropenia, decreased deformability, and delayed apoptosis. Rapid depletion of neonatal marrow PMN reserves during infection176 can lead to neutropenia with consequent impaired antimicrobial defenses and significantly increased risk for death.177 Neutropenia is particularly common in gram-negative sepsis in neonates.178 Release of immature neutrophil forms (bands), which have even greater dysfunction than mature neonatal neutrophils,179 can further predispose to adverse outcomes. PMN respiratory burst activity is also suppressed during sepsis and may contribute to poor microbicidal activity.180182 PMNs of neonates have reduced deformability compared with PMNs of adults, which, combined with the low blood pressure/flow state associated with septic shock, increases the risk of microvascular occlusion.174,183 Irreversible aggregation of newborn PMNs in the vascular space leads to decreased diapedesis, rapid depletion of bone marrow reserves, vascular crowding,183 and increased likelihood of compromised tissue perfusion184 leading to organ dysfunction. Neutrophils, although essential for combating pathogens, can also cause significant tissue damage and thus play a role in progression from sepsis to multi-organ system dysfunction. Reactive oxygen and nitrogen intermediates and proteolytic enzymes produced by PMNs can be released extracellularly, via activation of membrane-associated NADPH oxidase. Extracellular release of these reactive intermediates and enzymes can lead to destruction of nonphagocytized bacteria but can also cause local tissue destruction.185 Increased levels of neutrophil elastase as well as the neutrophil activators urokinase plasminogen activator, and urokinase plasminogen activator receptor have been described in infected neonates.109 Compared with adult PMNs, neonatal PMNs exhibit delayed apoptosis186,187 as well as sustained capacity for activation (CD11b upregulation) and cytotoxic function (ROI production) that contributes to tissue damage.188 Neutrophil-mediated damage may include endothelial and lung injury (including surfactant inactivation189) (see Fig. 2) in addition to other organ dysfunction (see Fig. 3).
Other Innate Cellular Contributions to Sepsis

Many other cells besides neutrophils are involved in the development of an immune response to infection, but the role that these cells play in the development of neonatal

454

Wynn & Wong

septic shock is incompletely characterized. Monocytes, macrophages, and dendritic cells amplify cellular recruitment through production of inflammatory mediators, phagocytosis and killing of pathogens, and antigen presentation to cells of the adaptive immune system. Important substances produced by stimulated monocytes that may contribute to septic shock include complement components, cytokines (proand antiinflammatory), coagulation factors, and extracellular matrix proteins (see Fig. 1).190 The role of NK cells in neonatal bacterial sepsis is incompletely defined. Despite activation,191 NK cytotoxicity is deficient in sepsis and recurrent infections.192,193 Circulating NK cells are decreased with neonatal shock.194 Further studies are necessary to more clearly define the role of NK cells in neonatal sepsis and shock. Mast cells play a role in the response to pathogen invasion via production of histamines (which promote vasodilation and upregulation of P-selectin) and cytokines (TNF-a, IL-1a/b), and by promoting neutrophil recruitment, direct bacterial phagocytosis, and antigen presentation.195 The production of histamine by mast cells likely contributes to the vasodilation associated with septic shock. Like eosinophils and PMNs, mast cells of adults are also capable of bacterial killing via generation of extracellular traps, like the neutrophil extracellular traps described previously.196 This means of immune protection has not been investigated in neonates. Mast cells may also alter adaptive immune function by patterning the TH2 immunosuppressive phenotype seen in the neonate and therefore contribute to the increased risk of infection. Immature dendritic cells exposed to histamine and LPS during maturation exhibit altered T-cell polarizing activity with predominance of TH2 phenotype via increased production of IL-10 and decreased production of IL-12.197 Furthermore, compared with mast cells of adults198 stimulated mast cells from neonates secrete significantly more histamine, which may contribute to vasodilation and the development of shock.199
Role of the Endothelium and Vasoactive Mediators in Septic Shock

Vascular endothelium has not historically been considered part of the innate cellular defenses, but recent studies have shown the importance of these sentinel cells in the early recognition and containment of microbial invasion. The endothelium can be a 2-edged sword, however, as excessive activation can lead to vascular dilation and leak, which are a driving forces behind the severe consequences of septic shock (see Fig. 3).124,200 Expression of TLRs allows endothelium to become activated in the presence of microbial components, leading to production of cytokines, chemokines, and adhesion molecules that attract circulating leukocytes and facilitate adherence.124 Vasoactive substances released from activated leukocytes, platelets, and endothelial cells are shown in Fig. 2 and include platelet-activating factor (PAF), thromboxane (TBX), leukotrienes (LTE), nitric oxide (NO), histamine, bradykinin, and prostaglandins (PGE).201,202 Activated PMNs produce phospholipase A2 (PLA2), which is increased in the serum of neonates with sepsis203 and leads to generation of vasoactive substances including PGE and LTE. Thromboxane produced by activated platelets and endothelin produced by activated endothelium204 are potent vasoconstrictors that participate in the development of pulmonary hypertension (PPHN).205208 Systemic overproduction of cytokines and vasoactive substances is associated with circulatory alterations and organ failure seen in severe sepsis and septic shock (see Fig. 3).25,209212 For example, the balance of NO and endothelin-1 (ET-1) may be disrupted with endothelial damage, favoring the constrictive effects of ET-1 leading to ischemia and injury. This may explain in part why NO inhibitors increased mortality in adults with septic shock.213

Treatment of Septic Shock in Neonates

455

Activated or damaged endothelium establishes a prothrombotic environment that can result in local microvascular occlusion165 or progress to DIC.214 Endothelial cell apoptosis, detachment from the lamina, and alterations in vascular tone combine to promote capillary leak of proteins and fluid leading to hypovolemia and shock.215 The role of endothelial activation during sepsis and septic shock in neonates, particularly in the premature infant, has not been thoroughly investigated. Adhesion molecules E- and P-selectin, expressed and secreted by activated endothelium, are increased in the serum of septic neonates109 and likely reflect significant endothelial activation. Toxins from GBS have been shown to damage pulmonary endothelium216 and likely participate in pulmonary complications associated with GBS pneumonia such as acute respiratory distress syndrome (ARDS) and PPHN.217 Using transgenic mice, it was recently shown that pulmonary endothelial cells sense blood-borne bacteria and their products,124 whereas alveolar macrophages patrol the airspaces for pathogens.218 These data help to explain in part the occurrence of ARDS and PPHN associated with severe sepsis in the absence of a primary pulmonary infectious focus.

PATHOPHYSIOLOGY OF SEPTIC SHOCK: CARDIOVASCULAR AND OTHER ORGAN EFFECTS Cardiovascular Effects

The hemodynamic response to sepsis has been less well characterized in premature and term neonates compared with children and adults, and the hemodynamic abnormalities are significantly more variable.219 Factors contributing to developmental differences in hemodynamic responses include altered structure and function of cardiomyocytes, limited ability to increase stroke volume and contractility, and contributions of the transition from fetal to neonatal circulation.220 A patent ductus arteriosus (PDA) and the presence of PPHN are significant modifying factors for the management of hypotension and hypoxia. In preterm infants with a PDA, aggressive volume administration to treat low blood pressure may lead to fluid overload, pulmonary edema, or heart failure. In the term infant with severe PPHN, on the other hand, aggressive volume and vasoactive medication administration to maintain a normal blood pressure may be beneficial by reducing right to left shunting and improving oxygenation. Although cardiomyopathy and heart failure may occasionally complicate sepsis in neonates, underlying coronary artery disease or other chronic cardiac conditions often present in septic adults do not complicate septic shock in the neonate. In adults, septic shock is most commonly characterized by reduced systemic vascular resistance and increased cardiac index.221 In children, a nonhyperdynamic state with reduced cardiac output and increased systemic vascular resistance is most common.219,222224 The hemodynamic presentation in neonates is much more variable219 and complicated by an unclear association between a normal blood pressure and adequate systemic blood flow.225,226 Abnormal peripheral vasoregulation with or with out myocardial dysfunction are the primary mechanisms for the hypotension accompanying septic shock in the neonate.227 Neonates with sepsis may present with tachycardia, poor perfusion and normal blood pressure (high systemic vascular resistance) or with hypotension and either adequate perfusion (warm shock, vasodilation) or inadequate perfusion (cold shock, vasoconstriction). These distinctions may be important for directing appropriate therapy to restore tissue perfusion, as discussed later.

456

Wynn & Wong

Multi-organ Dysfunction Syndrome

Septic shock that leads to multi-organ failure or MODS carries a dismal prognosis. Poor cardiac output and microcirculatory failure, sometimes combined with formation of microthrombi and DIC, can lead to compromised perfusion to the kidney,228,229 liver,230 gut,231 and central nervous system232 (see Fig. 3).59,210,233,234 Recent studies suggest that the mechanism of organ failure in sepsis may relate to decreased oxygen use associated with mitochondrial dysfunction rather than or in addition to poor oxygen delivery to tissues.235,236 Many other organ systems can be compromised in the setting of septic shock. Pulmonary complications include ARDS,237 secondary surfactant deficiency,238 pulmonary edema, pneumonia,23 and PPHN.220,237 Endocrine abnormalities may include adrenal insufficiency associated with refractory hypotension239 and altered thyroid function.240 Lymphocyte loss secondary to thymic involution and splenocyte apoptosis may also be present and may lead to a state of immune compromise following the acute phase of sepsis.241246 The importance of this finding has been shown in infected adults,247249 but the effect in neonates in whom adaptive immune function is immature is unknown. In a transgenic mouse model, neonatal animals lacking an adaptive immune system showed no difference in survival with polymicrobial sepsis compared with wild-type controls. This is in stark contrast to findings in adult mice.250 Hematologic findings during severe sepsis may include thrombocytopenia,170 neutropenia,177 and coagulation abnormalities including DIC.162 Sepsis can lead to metabolic and nutritional consequences. Increased energy expenditure and oxygen consumption251 and decreased mitochondrial oxidative function precipitated by hypoxia and the presence of damaging free radicals may lead to impaired growth and energy failure.252,253 The importance of providing optimum nutritional support in septic adults and children is increasingly recognized and should also be considered in septic neonates.

TREATMENT OF SEPSIS AND SEPTIC SHOCK Initial Resuscitation

Treatment guidelines for the management of severe sepsis and septic shock have been established for adults,254 children, and term neonates,255 but no such consensus guidelines exist for preterm neonates. The authors have attempted to incorporate the special circumstances related to premature physiology into the framework of treatment guidelines for term infants (Fig. 4). Development, testing, and acceptance of consensus guidelines for classification and management of preterm neonates with sepsis and septic shock are urgently needed to more systematically assess, diagnose, and treat these conditions. As with all emergencies in neonatology, management of septic shock begins with airway, breathing, and circulation. Septic neonates often present with apnea or severe respiratory distress and may require intubation.3,4 Following establishment of a secure airway and maintenance of lung volume for adequate gas exchange, administration of antibiotics and continuing assessment for cardiovascular dysfunction is critical. Shortly after birth, an umbilical vein catheter can be used for resuscitation but beyond this time, other peripheral or central venous access is essential for volume resuscitation, antibiotic administration, and pressor therapy. Timely therapy, including rapid restoration of adequate tissue perfusion, has been shown to improve outcomes in adults and children with sepsis, and should be the goal in neonates.

Treatment of Septic Shock in Neonates

Fig. 4. American College of Critical Care Medicine consensus guidelines for treatment of shock in term infants and suggested modifications for preterm infants. CI, cardiac index; CVP, central venous pressure; MAP, mean arterial pressure; NRP, Neonatal Resuscitation Program; PDA, patent ductus arteriosus; PPHN, persistent pulmonary hypertension of the newborn; RDS, respiratory distress syndrome; ScvO2, central venous oxygen saturation; SVC, superior vena cava; VLBW, very low birth weight. (From Brierley J, Carcillo JA, Choong K, et al. Clinical practice parameters for hemodynamic support of pediatric and neonatal septic shock: 2007 update from the American College of Critical Care Medicine. Crit Care Med 2009 Feb;37:66688; with permission.)

457

458

Wynn & Wong

Therapeutic Endpoints

In the absence of widely available or well-tested methods for quantifying hemodynamic compromise in septic shock in neonates, clinicians generally rely on vital signs and physical examination for decisions about therapy. Although mean arterial pressure (MAP) may not reflect systemic blood flow, monitoring blood pressure and other measures such as capillary refill time and urine output provide indirect information on the adequacy of organ blood flow. Suggestions for cardiovascular therapeutic end points in term neonates include a capillary refill time of less than 2 seconds, normal pulses without differential between peripheral and central pulses, warm extremities, urine output greater than 1 ml/kg/h, low serum lactate, and mixed venous saturation of more than 70%.256 Therapeutic end points in premature neonates have not been established but the goals for term infants seem reasonable. ELBW infants present the greatest challenge for determination of therapeutic end points in septic shock. Assessment of MAP, urine output, and capillary refill may not be particularly useful determinates of systemic blood flow in ELBW infants, particularly in the first 72 hours of life.257 In addition, the contribution of fetal hemoglobin may complicate accurate determination of central venous oxygen saturation (ScvO2) in neonates. ScvO2 obtained using hemoglobin A calibration is 4% to 7% higher compared with ScvO2 that accounts for fetal hemoglobin258 implying that perhaps the goal ScvO2 should be different in neonates than in older patients for optimum tissue oxygen delivery. In the future, monitoring techniques such as functional echocardiography (FE) and near-infrared spectroscopy (NIRS) may provide physiologic data to optimize management of septic shock. FE provides a bedside means to assess cardiac output, peripheral vascular resistance, and organ blood flow in response to volume, colloid, and vasoactive medications.259,260 FE can also be used to assess superior vena cava (SVC) flow, which has been suggested as a surrogate marker for cerebral blood flow261 and should be maintained at 40 ml/kg/min or higher.262 Prolonged decreases in SVC flow are associated with impaired neurodevelopmental outcome in very preterm neonates.263 In the absence of FE to monitor SVC flow, a capillary refill time of more than 4 seconds combined with a serum lactate concentration of more than 4 mmol/L had a specificity of 97% for identifying VLBW infants with a low SVC flow state on the first day of life.264 NIRS can be used to monitor end-organ perfusion noninvasively265 and is used often in neonates with congenital heart disease.266 A combination of FE and NIRS, in conjunction with traditional measures (MAP, SpO2, capillary refill, urine output) as well as intermittent laboratory evaluations of tissue perfusion such as pH, mixed venous saturation, lactate, and base deficit would be ideal for monitoring severity of septic shock and response to therapy.
Management of Hypotension and Cardiovascular Support

An algorithm for time-sensitive, goal-directed stepwise management of hemodynamic support for the term newborn with septic shock has been established and should be followed.255 Preterm neonates require specific caveats to this algorithm because of their unique physiology and risk for complications (see Fig. 4). In contrast to term neonates, the definition of hypotension and shock in preterm neonates is less clear, particularly in the immediate newborn period.26 Blood pressure may be a poor indicator of systemic blood flow in preterm neonates,225 yet objective measures of adequacy of tissue perfusion and oxygenation delivery are lacking. Another confounding variable in the management of neonatal shock is that inotrope use (dopamine, dobutamine) in hypotensive preterm neonates has not been shown to significantly improve short- or long-term outcomes.227,267,268 These

Treatment of Septic Shock in Neonates

459

considerations notwithstanding, and in absence of evidence of harm, some neonatologists advocate treating hypotension in preterm neonates to achieve a MAP of greater than or equal to 30 mm Hg. This goal MAP is based in part on a small study showing improved cerebral blood flow autoregulation above this threshold.269 However, a gestational age-based cutoff for normal blood pressure (goal MAP > GA) is used at many tertiary centers, especially in the first 3 days after birth. Clearly, more studies are required to determine whether targeting a specific blood pressure improves outcomes in preterm infants. Once a decision is made to treat hypotension with or without shock in a neonate, the recommended initial step is a fluid bolus (crystalloid). Although there is less data in neonates to support this intervention, it remains the accepted clinical practice to treat and monitor closely for signs of intravascular volume depletion.227 In term infants or older preterm infants, aggressive volume expansion (2040 mL/kg) should be considered. In contrast to outcomes with early aggressive fluid resuscitation in older populations,270 there is insufficient evidence to support early volume expansion in very preterm neonates,271 and there is a significant risk of intracranial hemorrhage associated with rapid volume expansion in the first few days after birth.272 In hypotensive preterm neonates, it is recommended that a single bolus of saline (1020 ml/kg over 3060 minutes) be given and if further intervention is necessary to begin vasoactive medications.268 In cases of obvious acute volume loss in preterm infants, more volume may be needed. Dopamine is generally the first-line vasoactive drug, with a starting dose of 5 to 10 mg/kg/min227 and dose escalation as needed. For neonates with shock, which is unresolved with volume resuscitation and dopamine, several possibilities exist for additional therapy, including glucocorticoids (see later discussion), other catecholamines, and inotropes/vasodilators. Epinephrine or norepinephrine infusions for refractory shock in neonates have been studied to a limited extent. Neonates with vasodilatory shock may have a positive response to the a-adrenergic vasoconstrictive effect of these agents. A recent report in term neonates showed the addition of noradrenaline to existing therapy (after fluid loading and dopamine or dobutamine infusion) resulted in increased blood pressure and decreased tissue lactate.18 In another study, low-dose epinephrine was found to be as effective as low-/ moderate-dose dopamine for increasing blood pressure, cerebral blood volume, and cerebral oxygen delivery in VLBW infants.273 Patients with depressed myocardial function may benefit from infusion of dobutamine for inotropy and vasodilation. In a study of 42 preterm neonates with low systemic blood flow (as determined by low SVC flow262) in the first 24 hours after birth, dobutamine treatment improved and maintained systemic blood flow better than dopamine.274,275 As a caution, dobutamine, particularly in high doses, can increase myocardial oxygen demand caused by b1 adrenergic stimulation. Dobutamine also has chronotropic actions and severe tachycardia may lead to decreased cardiac output that may be corrected by decreasing the dose. Milrinone, a phosphodiesterase inhibitor and inodilator, has not been studied in neonatal septic shock but has been used in pediatric patients with septic shock.276,277 In a study of patients aged 9 months to 15 years with volume-resuscitated catecholamine-resistant nonhyperdynamic septic shock, milrinone increased cardiac index, stroke volume, and oxygen delivery, and decreased systemic vascular resistance without increasing heart rate or blood pressure.276 Another alternative agent for treating septic shock is the vasoconstrictor argininevasopressin (AVP) or its longer half-life analogue terlipressin.278 In a report of 6 ELBW infants, AVP improved MAP and urine output in patients with septic shock but not in those with nonseptic shock.279

460

Wynn & Wong

Hydrocortisone Treatment in Neonatal Septic Shock

Induced by proinflammatory cytokines, endogenous cortisol attenuates the intensity of the systemic inflammatory response associated with severe sepsis and septic shock.280 Studies in adults have shown that high-dose glucocorticoid therapy does not affect sepsis mortality although low-dose therapy may be beneficial.254 In 1 randomized clinical trial, low-dose cortisol treatment in conjunction with standard of care measures was associated with a reduction in mortality in adults with septic shock and adrenal insufficiency.281 In another study in adults, cortisol treatment sped the reversal of septic shock but had no effect on mortality.282 Cortisol production in the neonate is significantly increased early in septic shock.283 However, very preterm neonates can have relative adrenal insufficiency that may contribute to hemodynamic instability and hypotension. In many clinical practices, hydrocortisone is the third-line agent in treatment of neonatal shock after volume resuscitation and dopamine.227,268,284 In addition to its cytokine-suppressing effects, hydrocortisone has been shown to increase the sensitivity of the cardiovascular system to endogenous or exogenous catecholamines, resulting in improvements in myocardial contractility, stroke volume, effective circulating blood volume, systemic vascular resistance, and urine output. Hydrocortisone has not been evaluated in prospective randomized clinical trials for the treatment of septic shock in the neonate, but it has been shown to increase blood pressure, decrease heart rate, and decrease vasoactive medication requirements in preterm and term neonates.284,285 If hydrocortisone treatment is considered, obtaining a pretreatment serum cortisol level is prudent to differentiate contributing causes of hypotension. The reader is referred to a recent review on the diagnosis and treatment of adrenal insufficiency in the premature neonate.286
Pulmonary Support

Increased inspired oxygen may be necessary in the setting of neonatal septic shock to maximize tissue oxygen delivery. Decreased pulmonary function (RDS) and/or respiratory failure (apnea) in conjunction with increased tissue demand (increased respiratory and metabolic activity associated with acidosis) contribute to tissue hypoxia. Mechanical ventilation can improve gas exchange through maintenance of lung volume and decreased work of breathing. Administration of exogenous surfactant to neonates with severe pneumonia has been shown to improve oxygenation and gas exchange and reduce the need for extracorporeal membrane oxygenation (ECMO).238In extremely sick neonates, consideration should be given to maintaining a normal or near-normal pH and oxygen saturations in the 90s rather than allowing permissive hypercapnia and lower saturations, which is standard practice in healthy preterm neonates. Normalizing pH and arterial oxygen content may improve cardiac contractility and improve tissue oxygen content, thus decreasing the risk of multiorgan dysfunction and the risk of pulmonary hypertension. Infants with sepsis and PPHN may require inhaled nitric oxide (iNO) in addition to optimized ventilation strategies such as high frequency oscillatory ventilation.287 If oxygenation or tissue perfusion remain severely compromised despite optimal medical management, ECMO should be considered in neonates greater than 2 kg without contraindications such as the presence of or high risk for acute hemorrhage.288,289
OTHER SUPPORTIVE CARE OF NEONATES WITH SEPTIC SHOCK

Avoidance of hypothermia and hypoglycemia is important in neonates with septic shock. With the exception of patients with acute perinatal hypoxic ischemic

Treatment of Septic Shock in Neonates

461

encephalopathy,290 normothermia should be maintained on a radiant warmer. Use of a 10% glucose solution delivering 4 to 6 mg/kg/min of glucose combined with frequent monitoring to ensure normoglycemia is recommended. Correction of a significant coagulopathy and anemia (hemoglobin %10 g/dL) through the transfusion of fresh frozen plasma or packed red blood cells may also serve to improve blood pressure291 and oxygen delivery. The importance of providing adequate protein and calories to the infant with sepsis and septic shock cannot be overstated. Increased energy demands promote catabolism if adequate nutrition is not provided. Premature neonates have decreased muscles mass and energy reserves as well as higher baseline nutritional requirements compared with term neonates.292 Increase in serum triglyceride levels during sepsis293 and increased serum oxygen-derived free radicals associated with infusions of lipid have prompted some clinicians to withhold or decrease intralipid infusions. A recent study showed concurrent administration of intralipids in neonates with infection is not associated with hypertriglyceridemia in the absence of liver dysfunction or fetal growth restriction.294 It is suggested that intralipid infusions during sepsis or septic shock in neonates be accompanied by careful monitoring of serum triglycerides to avoid hypertriglyceridemia. Maintenance of a carbohydrate to lipid ratio of w3:1 increases fat use and decreases production of oxygen-derived free radicals to levels seen with fat exclusion.295 Protein intakes of 2 to 3 g/kg/d are generally not associated with azotemia, hyperammonemia, or metabolic acidosis296 in the setting of sepsis, but monitoring of blood urea nitrogen is recommended. Monitoring liver and renal function is important for assessing the effectiveness of therapies to improve tissue perfusion and for making decisions about dosing medications that require modification for elimination.
ALTERNATIVE IMMUNOLOGIC AND PHARMACOTHERAPIES FOR NEONATAL SEPSIS/SHOCK

There have been many attempts directed at improving outcomes of sepsis and septic shock in neonates via immunomodulation. A complete review of adjunct immunologic therapies in neonatal sepsis, see the article by Tarnow-Mordi and colleagues elsewhere in this issue for further exploration of this topic.
OUTCOMES WITH SEPSIS AND SEPTIC SHOCK

The outcome of septic shock in the neonate is dismal. One study reported death or severe sequelae in 52% of infants, with only 28% of infants less than 1000 g alive and free of disability at 18 months of age.7 Variables predictive of mortality include cardiac dysfunction manifested as refractory shock, acute renal failure, neutropenia, increased prothrombin time, excessive bleeding, metabolic acidosis, and hypothermia.231,297 Neurodevelopmental outcomes following neonatal sepsis, without stratification for shock, have been studied in some detail and demonstrate significant risk for impairment, particularly in the most premature neonates.298 VLBW infants with sepsis, compared with those without, have been reported to have significantly increased mortality (21% vs 9%), longer hospital stay (98 vs 58 days), and a higher risk of chronic lung disease.31 ELBW infants are at especially high risk for sepsis-associated adverse neurodevelopmental outcomes, including deafness, cerebral palsy, lower mental and psychomotor development scores, and vision impairment.299,300 In a study of preterm infants, white matter abnormality on magnetic resonance imaging at term corrected age-predicted neurodevelopmental impairment in those with sepsis compared with those without.301 Surgical NEC, which is often accompanied by SIRS or shock, has

462

Wynn & Wong

been associated with significant growth delay and adverse neurodevelopmental outcomes at 18 to 22 months.302 A study of ELBW infants with systemic candidiasis found that 73% died or developed a neurodevelopmental impairment63 including retinopathy.303 These data show that the toll of neonatal sepsis and septic shock reaches far beyond the acute complications of organ dysfunction and mortality.
FUTURE CONSIDERATIONS

Neonatal sepsis requires translational and clinical research. Definitions for the sepsis continuum and treatment algorithms specific for preterm infants should be developed to improve the quality of clinical trials and facilitate meta-analyses of prophylactic and therapeutic interventions. Systems biology and genomic and proteomic studies have yielded important data on septic shock in older populations304312 and the use of these modern techniques in the study of neonatal inflammation and response to pathogen challenge has begun.109,138,313 With further research, real-time sampling using only microliters of blood will allow rapid identification of highest-risk patients, pathogenspecific responses, and sepsis-staging biomarkers.314 Immaturities of immune function and physiology in the neonate necessitate developmental stage-specific evaluations of sepsis pathophysiology and treatment. Exploration of adjuvant treatments including LPS-binding proteins (rBPI,315 sCD14, or anti-CD14316), antiinflammatory therapies (pentoxifylline,317 nicotinic stimulation,318 statins319), synthetic host defense peptides (rhSP-D,320 lactoferrin321,322), combination therapies323 (ie, IVIg and colony-stimulating factor), and innate immune priming using TLR agonists250 may yield improved outcomes. Advances in these areas are urgently needed and are likely to substantially improve long-term outcomes.
SUMMARY

Neonatal septic shock is a devastating condition associated with high morbidity and mortality, Definitions for the sepsis continuum and treatment algorithms specific for premature neonates are needed to improve studies of septic shock and assess benefit from clinical interventions. Unique features of the immature immune system and pathophysiologic responses to sepsis, particularly those of extremely preterm infants, necessitate that clinical trials consider them as a separate group. Keen clinical suspicion and knowledge of risk factors will help to identify those neonates at greatest risk for development of septic shock. Genomic and proteomic approaches, particularly those that use very small sample volumes, will increase our understanding of the pathophysiology and direct the development of novel agents for prevention and treatment of severe sepsis and shock in the neonate. Although at present antimicrobial therapy and supportive care remain the foundation of treatment, in the future immunomodulatory agents are likely to improve outcomes for this vulnerable population.
ACKNOWLEDGMENTS

The authors thank Associate Professor C. Michael Cotten, MD, MHS for his review of this manuscript.
REFERENCES

1. Lawn JE, Cousens S, Zupan J. 4 million neonatal deaths: when? Where? Why? Lancet 2005;365(9462):891900. 2. Stoll BJ, Hansen N, Fanaroff AA, et al. Changes in pathogens causing earlyonset sepsis in very-low-birth-weight infants. N Engl J Med 2002;347(4):2407.

Treatment of Septic Shock in Neonates

463

3. Stoll BJ, Hansen N, Fanaroff AA, et al. Late-onset sepsis in very low birth weight neonates: the experience of the NICHD Neonatal Research Network. Pediatrics 2002;110(2 Pt 1):28591. 4. Stoll BJ, Hansen NI, Higgins RD, et al. Very low birth weight preterm infants with early onset neonatal sepsis: the predominance of Gram-negative infections continues in the National Institute of Child Health and Human Development Neonatal Research Network, 20022003. Pediatr Infect Dis J 2005; 24(7):6359. 5. Haque KN, Khan MA, Kerry S, et al. Pattern of culture-proven neonatal sepsis in a district general hospital in the United Kingdom. Infect Control Hosp Epidemiol 2004;25(9):75964. 6. Haque KN. Defining common infections in children and neonates. J Hosp Infect 2007;65(Suppl 2):1104. 7. Kermorvant-Duchemin E, Laborie S, Rabilloud M, et al. Outcome and prognostic factors in neonates with septic shock. Pediatr Crit Care Med 2008;9(2):18691. 8. Furman WL, Menke JA, Barson WJ, et al. Continuous naloxone infusion in two neonates with septic shock. J Pediatr 1984;105(4):64951. 9. Togari H, Mikawa M, Iwanaga T, et al. Endotoxin clearance by exchange blood transfusion in septic shock neonates. Acta Paediatr Scand 1983;72(1):8791. 10. Fenton LJ, Strunk RC. Complement activation and group B streptococcal infection in the newborn: similarities to endotoxin shock. Pediatrics 1977;60(6):9017. 11. Tollner U, Pohlandt F. Septicemia in the newborn due to Gram-negative bacilli. Risk factors, clinical symptoms, and hematologic changes. Eur J Pediatr 1976;123(4):24354. 12. Frommhold D, Birle A, Linderkamp O, et al. Drotrecogin alpha (activated) in neonatal septic shock. Scand J Infect Dis 2005;37(4):3068. 13. Miyairi I, Berlingieri D, Protic J, et al. Neonatal invasive group A streptococcal disease: case report and review of the literature. Pediatr Infect Dis J 2004; 23(2):1615. 14. Ahmed K, Sein PP, Shahnawaz M, et al. Pasteurella gallinarum neonatal meningitis. Clin Microbiol Infect 2002;8(1):557. 15. Roll C, Schmid EN, Menken U, et al. Fatal Salmonella enteritidis sepsis acquired prenatally in a premature infant. Obstet Gynecol 1996;88(4 Pt 2):6923. 16. Wolfler A, Silvani P, Musicco M, et al. Incidence of and mortality due to sepsis, severe sepsis and septic shock in Italian Pediatric Intensive Care Units: a prospective national survey. Intensive Care Med 2008;34(9):16907. 17. Rodriguez-Nunez A, Lopez-Herce J, Gil-Anton J, et al. Rescue treatment with terlipressin in children with refractory septic shock: a clinical study. Crit Care 2006;10(1):R20. 18. Tourneux P, Rakza T, Abazine A, et al. Noradrenaline for management of septic shock refractory to fluid loading and dopamine or dobutamine in full-term newborn infants. Acta Paediatr 2008;97(2):17780. 19. Filippi L, Poggi C, Serafini L, et al. Terlipressin as rescue treatment of refractory shock in a neonate. Acta Paediatr 2008;97(4):5002. 20. Meyer S, Loffler G, Polcher T, et al. Vasopressin in catecholamine-resistant septic and cardiogenic shock in very-low-birthweight infants. Acta Paediatr 2006;95(10):130912. 21. McAdams RM, Garza-Cox S, Yoder BA. Early-onset neonatal pneumococcal sepsis syndrome. Pediatr Crit Care Med 2005;6(5):5957. 22. Matok I, Leibovitch L, Vardi A, et al. Terlipressin as rescue therapy for intractable hypotension during neonatal septic shock. Pediatr Crit Care Med 2004;5(2):1168.

464

Wynn & Wong

23. Aikio O, Vuopala K, Pokela ML, et al. Diminished inducible nitric oxide synthase expression in fulminant early-onset neonatal pneumonia. Pediatrics 2000; 105(5):10139. 24. Duke TD, Butt W, South M. Predictors of mortality and multiple organ failure in children with sepsis. Intensive Care Med 1997;23(6):68492. 25. Goldstein B, Giroir B, Randolph A. International pediatric sepsis consensus conference: definitions for sepsis and organ dysfunction in pediatrics. Pediatr Crit Care Med 2005;6(1):28. 26. Cayabyab R, McLean CW, Seri I. Definition of hypotension and assessment of hemodynamics in the preterm neonate. J Perinatol 2009;29(Suppl 2):S5862. 27. Shah GS, Budhathoki S, Das BK, et al. Risk factors in early neonatal sepsis. Kathmandu Univ Med J (KUMJ) 2006;4(2):18791. 28. Salem SY, Sheiner E, Zmora E, et al. Risk factors for early neonatal sepsis. Arch Gynecol Obstet 2006;274(4):198202. 29. Yancey MK, Duff P, Kubilis P, et al. Risk factors for neonatal sepsis. Obstet Gynecol 1996;87(2):18894. 30. Benitz WE, Gould JB, Druzin ML. Risk factors for early-onset group B streptococcal sepsis: estimation of odds ratios by critical literature review. Pediatrics 1999;103(6):e77. 31. Fanaroff AA, Korones SB, Wright LL, et al. Incidence, presenting features, risk factors and significance of late onset septicemia in very low birth weight infants. The National Institute of Child Health and Human Development Neonatal Research Network. Pediatr Infect Dis J 1998;17(7):5938. 32. Schuchat A, Zywicki SS, Dinsmoor MJ, et al. Risk factors and opportunities for prevention of early-onset neonatal sepsis: a multicenter case-control study. Pediatrics 2000;105(1 Pt 1):216. 33. Escobar GJ, Li DK, Armstrong MA, et al. Neonatal sepsis workups in infants >/52000 grams at birth: a population-based study. Pediatrics 2000;106(2 Pt 1):25663. 34. Sharma R, Tepas JJ 3rd, Hudak ML, et al. Neonatal gut barrier and multiple organ failure: role of endotoxin and proinflammatory cytokines in sepsis and necrotizing enterocolitis. J Pediatr Surg 2007;42(3):45461. 35. Sonntag J, Wagner MH, Waldschmidt J, et al. Multisystem organ failure and capillary leak syndrome in severe necrotizing enterocolitis of very low birth weight infants. J Pediatr Surg 1998;33(3):4814. 36. Dahmer MK, Randolph A, Vitali S, et al. Genetic polymorphisms in sepsis. Pediatr Crit Care Med 2005;6(Suppl 3):S6173. 37. Cogulu O, Onay H, Uzunkaya D, et al. Role of angiotensin-converting enzyme gene polymorphisms in children with sepsis and septic shock. Pediatr Int 2008;50(4):47780. 38. Lyons EJ, Amos W, Berkley JA, et al. Homozygosity and risk of childhood death due to invasive bacterial disease. BMC Med Genet 2009;10:55. 39. Liangos O, Jaber BL. Multiple organ dysfunction syndrome in children with sepsis: role of genetic factors. Semin Nephrol 2008;28(5):499509. 40. Kumpf O, Schumann RR. Genetic influence on bloodstream infections and sepsis. Int J Antimicrob Agents 2008;32(Suppl 1):S4450. 41. Sutherland AM, Walley KR. Bench-to-bedside review: association of genetic variation with sepsis. Crit Care 2009;13(2):210. 42. Ahrens P, Kattner E, Kohler B, et al. Mutations of genes involved in the innate immune system as predictors of sepsis in very low birth weight infants. Pediatr Res 2004;55(4):6526.

Treatment of Septic Shock in Neonates

465

43. Baier RJ, Loggins J, Yanamandra K. IL-10, IL-6 and CD14 polymorphisms and sepsis outcome in ventilated very low birth weight infants. BMC Med 2006;4:10. 44. Chauhan M, McGuire W. Interleukin-6 (-174C) polymorphism and the risk of sepsis in very low birth weight infants: meta-analysis. Arch Dis Child Fetal Neonatal Ed 2008;93(6):F4279. 45. Dzwonek AB, Neth OW, Thiebaut R, et al. The role of mannose-binding lectin in susceptibility to infection in preterm neonates. Pediatr Res 2008;63(6):6805. 46. Gopel W, Hartel C, Ahrens P, et al. Interleukin-6-174-genotype, sepsis and cerebral injury in very low birth weight infants. Genes Immun 2006;7(1):658. 47. Hartel C, Rupp J, Hoegemann A, et al. 159C>T CD14 genotypefunctional effects on innate immune responses in term neonates. Hum Immunol 2008; 69(6):33843. 48. Hartel C, Schultz C, Herting E, et al. Genetic association studies in VLBW infants exemplifying susceptibility to sepsisrecent findings and implications for future research. Acta Paediatr 2007;96(2):15865. 49. Hubacek JA, Stuber F, Frohlich D, et al. Gene variants of the bactericidal/permeability increasing protein and lipopolysaccharide binding protein in sepsis patients: gender-specific genetic predisposition to sepsis. Crit Care Med 2001;29(3):55761. 50. Mollen KP, Gribar SC, Anand RJ, et al. Increased expression and internalization of the endotoxin coreceptor CD14 in enterocytes occur as an early event in the development of experimental necrotizing enterocolitis. J Pediatr Surg 2008; 43(6):117581. 51. Reiman M, Kujari H, Ekholm E, et al. Interleukin-6 polymorphism is associated with chorioamnionitis and neonatal infections in preterm infants. J Pediatr 2008;153(1):1924. 52. Schueller AC, Heep A, Kattner E, et al. Prevalence of two tumor necrosis factor gene polymorphisms in premature infants with early onset sepsis. Biol Neonate 2006;90(4):22932. 53. Treszl A, Heninger E, Kalman A, et al. Lower prevalence of IL-4 receptor alphachain gene G variant in very-low-birth-weight infants with necrotizing enterocolitis. J Pediatr Surg 2003;38(9):13748. 54. Treszl A, Kocsis I, Szathmari M, et al. Genetic variants of TNF-[FC12]a, IL-1beta, IL-4 receptor [FC12]a-chain, IL-6 and IL-10 genes are not risk factors for sepsis in low-birth-weight infants. Biol Neonate 2003;83(4):2415. 55. Treszl A, Kocsis I, Szathmari M, et al. Genetic variants of the tumour necrosis factor-alpha promoter gene do not influence the development of necrotizing enterocolitis. Acta Paediatr 2001;90(10):11825. 56. Treszl A, Tulassay T, Vasarhelyi B. Genetic basis for necrotizing enterocolitisrisk factors and their relations to genetic polymorphisms. Front Biosci 2006;11: 57080. 57. Verboon-Maciolek MA, Krediet TG, Gerards LJ, et al. Clinical and epidemiologic characteristics of viral infections in a neonatal intensive care unit during a 12year period. Pediatr Infect Dis J 2005;24(10):9014. 58. Verboon-Maciolek MA, Krediet TG, Gerards LJ, et al. Severe neonatal parechovirus infection and similarity with enterovirus infection. Pediatr Infect Dis J 2008; 27(3):2415. 59. Kawada J, Kimura H, Ito Y, et al. Evaluation of systemic inflammatory responses in neonates with herpes simplex virus infection. J Infect Dis 2004; 190(3):4948.

466

Wynn & Wong

60. Karlowicz MG, Buescher ES, Surka AE. Fulminant late-onset sepsis in a neonatal intensive care unit, 19881997, and the impact of avoiding empiric vancomycin therapy. Pediatrics 2000;106(6):138790. 61. Hyde TB, Hilger TM, Reingold A, et al. Trends in incidence and antimicrobial resistance of early-onset sepsis: population-based surveillance in San Francisco and Atlanta. Pediatrics 2002;110(4):6905. 62. Benjamin DK, DeLong E, Cotten CM, et al. Mortality following blood culture in premature infants: increased with Gram-negative bacteremia and candidemia, but not Gram-positive bacteremia. J Perinatol 2004;24(3):17580. 63. Benjamin DK Jr, Stoll BJ, Fanaroff AA, et al. Neonatal candidiasis among extremely low birth weight infants: risk factors, mortality rates, and neurodevelopmental outcomes at 18 to 22 months. Pediatrics 2006;117(1):8492. 64. Schelonka RL, Chai MK, Yoder BA, et al. Volume of blood required to detect common neonatal pathogens. J Pediatr 1996;129(2):2758. 65. Kawai T, Akira S. The roles of TLRs, RLRs and NLRs in pathogen recognition. Int Immunol 2009;21(4):31737. 66. Kumagai Y, Takeuchi O, Akira S. Pathogen recognition by innate receptors. J Infect Chemother 2008;14(2):8692. 67. Trinchieri G, Sher A. Cooperation of Toll-like receptor signals in innate immune defence. Nat Rev Immunol 2007;7(3):17990. 68. Rittirsch D, Flierl MA, Ward PA. Harmful molecular mechanisms in sepsis. Nat Rev Immunol 2008;8(10):77687. 69. Krumbiegel D, Zepp F, Meyer CU. Combined Toll-like receptor agonists synergistically increase production of inflammatory cytokines in human neonatal dendritic cells. Hum Immunol 2007;68(10):81322. 70. Bochud PY, Chien JW, Marr KA, et al. Toll-like receptor 4 polymorphisms and aspergillosis in stem-cell transplantation. N Engl J Med 2008;359(17): 176677. 71. Wurfel MM, Gordon AC, Holden TD, et al. Toll-like receptor 1 polymorphisms affect innate immune responses and outcomes in sepsis. Am J Respir Crit Care Med 2008;178(7):71020. 72. Agnese DM, Calvano JE, Hahm SJ, et al. Human toll-like receptor 4 mutations but not CD14 polymorphisms are associated with an increased risk of Gramnegative infections. J Infect Dis 2002;186(10):15225. 73. Lorenz E, Mira JP, Cornish KL, et al. A novel polymorphism in the toll-like receptor 2 gene and its potential association with staphylococcal infection. Infect Immun 2000;68(11):6398401. 74. Zhang SY, Jouanguy E, Ugolini S, et al. TLR3 deficiency in patients with herpes simplex encephalitis. Science 2007;317(5844):15227. 75. Mockenhaupt FP, Cramer JP, Hamann L, et al. Toll-like receptor (TLR) polymorphisms in African children: common TLR-4 variants predispose to severe malaria. J Commun Dis 2006;38(3):23045. 76. Faber J, Meyer CU, Gemmer C, et al. Human toll-like receptor 4 mutations are associated with susceptibility to invasive meningococcal disease in infancy. Pediatr Infect Dis J 2006;25(1):801. 77. Zhang JP, Chen C, Yang Y. [Changes and clinical significance of Toll-like receptor 2 and 4 expression in neonatal infections]. Zhonghua Er Ke Za Zhi 2007;45(2):1303 [in Chinese]. 78. Leaphart CL, Cavallo J, Gribar SC, et al. A critical role for TLR4 in the pathogenesis of necrotizing enterocolitis by modulating intestinal injury and repair. J Immunol 2007;179(7):480820.

Treatment of Septic Shock in Neonates

467

79. Goldbach-Mansky R, Dailey NJ, Canna SW, et al. Neonatal-onset multisystem inflammatory disease responsive to interleukin-1beta inhibition. N Engl J Med 2006;355(6):58192. 80. Szebeni B, Szekeres R, Rusai K, et al. Genetic polymorphisms of CD14, toll-like receptor 4, and caspase-recruitment domain 15 are not associated with necrotizing enterocolitis in very low birth weight infants. J Pediatr Gastroenterol Nutr 2006;42(1):2731. 81. Shigemoto T, Kageyama M, Hirai R, et al. Identification of loss of function mutations in human genes encoding RIG-I and mda5: implications for resistance to type I diabetes. J Biol Chem 2009;284(20):1334854. 82. Warren SE, Mao DP, Rodriguez AE, et al. Multiple Nod-like receptors activate caspase 1 during Listeria monocytogenes infection. J Immunol 2008;180(11): 755864. 83. Behrendt D, Dembinski J, Heep A, et al. Lipopolysaccharide binding protein in preterm infants. Arch Dis Child Fetal Neonatal Ed 2004;89(6):F5514. 84. Berner R, Furll B, Stelter F, et al. Elevated levels of lipopolysaccharide-binding protein and soluble CD14 in plasma in neonatal early-onset sepsis. Clin Diagn Lab Immunol 2002;9(2):4405. 85. Blanco A, Solis G, Arranz E, et al. Serum levels of CD14 in neonatal sepsis by Gram-positive and Gram-negative bacteria. Acta Paediatr 1996;85(6):72832. 86. Gu W, Shan YA, Zhou J, et al. Functional significance of gene polymorphisms in the promoter of myeloid differentiation-2. Ann Surg 2007;246(1):1518. 87. von Bernuth H, Picard C, Jin Z, et al. Pyogenic bacterial infections in humans with MyD88 deficiency. Science 2008;321(5889):6916. 88. Picard C, Puel A, Bonnet M, et al. Pyogenic bacterial infections in humans with IRAK-4 deficiency. Science 2003;299(5615):20769. 89. Ku CL, Picard C, Erdos M, et al. IRAK4 and NEMO mutations in otherwise healthy children with recurrent invasive pneumococcal disease. J Med Genet 2007;44(1):1623. 90. Lotze MT, Tracey KJ. High-mobility group box 1 protein (HMGB1): nuclear weapon in the immune arsenal. Nat Rev Immunol 2005;5(4):33142. 91. Mullins GE, Sunden-Cullberg J, Johansson AS, et al. Activation of human umbilical vein endothelial cells leads to relocation and release of high-mobility group box chromosomal protein 1. Scand J Immunol 2004;60(6):56673. 92. van Zoelen MA, Yang H, Florquin S, et al. Role of toll-like receptors 2 and 4, and the receptor for advanced glycation end products in high-mobility group box 1induced inflammation in vivo. Shock 2009;31(3):2804. 93. Sappington PL, Yang R, Yang H, et al. HMGB1 B box increases the permeability of Caco-2 enterocytic monolayers and impairs intestinal barrier function in mice. Gastroenterology 2002;123(3):790802. 94. Zamora R, Grishin A, Wong C, et al. High-mobility group box 1 protein is an inflammatory mediator in necrotizing enterocolitis: protective effect of the macrophage deactivator semapimod. Am J Physiol Gastrointest Liver Physiol 2005;289(4):G64352. 95. Pack CD, Kumaraguru U, Suvas S, et al. Heat-shock protein 70 acts as an effective adjuvant in neonatal mice and confers protection against challenge with herpes simplex virus. Vaccine 2005;23(27):352634. 96. Wheeler DS, Lahni P, Odoms K, et al. Extracellular heat shock protein 60 (Hsp60) levels in children with septic shock. Inflamm Res 2007;56(5):2169. 97. Wheeler DS, Fisher LE Jr, Catravas JD, et al. Extracellular hsp70 levels in children with septic shock. Pediatr Crit Care Med 2005;6(3):30811.

468

Wynn & Wong

98. Kono H, Rock KL. How dying cells alert the immune system to danger. Nat Rev Immunol 2008;8(4):27989. 99. Batra S, Kumar R, Seema, et al. Alterations in antioxidant status during neonatal sepsis. Ann Trop Paediatr 2000;20(1):2733. 100. Kapoor K, Basu S, Das BK, et al. Lipid peroxidation and antioxidants in neonatal septicemia. J Trop Pediatr 2006;52(5):3725. 101. Ng PC. Diagnostic markers of infection in neonates. Arch Dis Child Fetal Neonatal Ed 2004;89(3):F22935. 102. Ng PC, Li K, Wong RP, et al. Proinflammatory and anti-inflammatory cytokine responses in preterm infants with systemic infections. Arch Dis Child Fetal Neonatal Ed 2003;88(3):F20913. 103. Levy O, Zarember KA, Roy RM, et al. Selective impairment of TLR-mediated innate immunity in human newborns: neonatal blood plasma reduces monocyte TNF-alpha induction by bacterial lipopeptides, lipopolysaccharide, and imiquimod, but preserves the response to R-848. J Immunol 2004;173(7): 462734. 104. Bozza FA, Salluh JI, Japiassu AM, et al. Cytokine profiles as markers of disease severity in sepsis: a multiplex analysis. Crit Care 2007;11(2):R49. 105. Hodge G, Hodge S, Haslam R, et al. Rapid simultaneous measurement of multiple cytokines using 100 microl sample volumesassociation with neonatal sepsis. Clin Exp Immunol 2004;137(2):4027. 106. Heper Y, Akalin EH, Mistik R, et al. Evaluation of serum C-reactive protein, procalcitonin, tumor necrosis factor alpha, and interleukin-10 levels as diagnostic and prognostic parameters in patients with community-acquired sepsis, severe sepsis, and septic shock. Eur J Clin Microbiol Infect Dis 2006;25(8): 48191. 107. Atici A, Satar M, Cetiner S, et al. Serum tumor necrosis factor-alpha in neonatal sepsis. Am J Perinatol 1997;14(7):4014. 108. Sadeghi K, Berger A, Langgartner M, et al. Immaturity of infection control in preterm and term newborns is associated with impaired toll-like receptor signaling. J Infect Dis 2007;195(2):296302. 109. Kingsmore SF, Kennedy N, Halliday HL, et al. Identification of diagnostic biomarkers for infection in premature neonates. Mol Cell Proteomics 2008; 7(10):186375. 110. Grobmyer SR, Lin E, Lowry SF, et al. Elevation of IL-18 in human sepsis. J Clin Immunol 2000;20(3):2125. 111. Hirata J, Kotani J, Aoyama M, et al. A role for IL-18 in human neutrophil apoptosis. Shock 2008;30(6):62833. 112. Cusumano V, Midiri A, Cusumano VV, et al. Interleukin-18 is an essential element in host resistance to experimental group B streptococcal disease in neonates. Infect Immun 2004;72(1):295300. 113. Puren AJ, Fantuzzi G, Gu Y, et al. Interleukin-18 (IFNgamma-inducing factor) induces IL-8 and IL-1beta via TNFalpha production from non-CD141 human blood mononuclear cells. J Clin Invest 1998;101(3):71121. 114. Elbim C, Guichard C, Dang PM, et al. Interleukin-18 primes the oxidative burst of neutrophils in response to formyl-peptides: role of cytochrome b558 translocation and N-formyl peptide receptor endocytosis. Clin Diagn Lab Immunol 2005; 12(3):43646. 115. Dollner H, Vatten L, Austgulen R. Early diagnostic markers for neonatal sepsis: comparing C-reactive protein, interleukin-6, soluble tumour necrosis factor receptors and soluble adhesion molecules. J Clin Epidemiol 2001;54(12):12517.

Treatment of Septic Shock in Neonates

469

116. Turunen R, Andersson S, Nupponen I, et al. Increased CD11b-density on circulating phagocytes as an early sign of late-onset sepsis in extremely low-birthweight infants. Pediatr Res 2005;57(2):2705. 117. Figueras-Aloy J, Gomez-Lopez L, Rodriguez-Miguelez JM, et al. Serum soluble ICAM-1, VCAM-1, L-selectin, and P-selectin levels as markers of infection and their relation to clinical severity in neonatal sepsis. Am J Perinatol 2007;24(6):3318. 118. Kourtis AP, Lee FK, Stoll BJ. Soluble L-selectin, a marker of immune activation, in neonatal infection. Clin Immunol 2003;109(2):2248. 119. McEvoy LT, Zakem-Cloud H, Tosi MF. Total cell content of CR3 (CD11b/CD18) and LFA-1 (CD11a/CD18) in neonatal neutrophils: relationship to gestational age. Blood 1996;87(9):392933. 120. Buhrer C, Graulich J, Stibenz D, et al. L-selectin is down-regulated in umbilical cord blood granulocytes and monocytes of newborn infants with acute bacterial infection. Pediatr Res 1994;36(6):799804. 121. Sullivan SE, Staba SL, Gersting JA, et al. Circulating concentrations of chemokines in cord blood, neonates, and adults. Pediatr Res 2002;51(5):6537. 122. Anderson DC, Rothlein R, Marlin SD, et al. Impaired transendothelial migration by neonatal neutrophils: abnormalities of Mac-1 (CD11b/CD18)-dependent adherence reactions. Blood 1990;76(12):261321. 123. Meade VM, Barese CN, Kim C, et al. Rac2 concentrations in umbilical cord neutrophils. Biol Neonate 2006;90(3):1569. 124. Hickey MJ, Kubes P. Intravascular immunity: the host-pathogen encounter in blood vessels. Nat Rev Immunol 2009;9(5):36475. 125. Ng PC, Li K, Chui KM, et al. IP-10 is an early diagnostic marker for identification of late-onset bacterial infection in preterm infants. Pediatr Res 2007;61(1):938. 126. Wong HR, Cvijanovich N, Wheeler DS, et al. Interleukin-8 as a stratification tool for interventional trials involving pediatric septic shock. Am J Respir Crit Care Med 2008;178(3):27682. 127. Ng PC, Li K, Leung TF, et al. Early prediction of sepsis-induced disseminated intravascular coagulation with interleukin-10, interleukin-6, and RANTES in preterm infants. Clin Chem 2006;52(6):11819. 128. Sriskandan S, Altmann DM. The immunology of sepsis. J Pathol 2008;214(2): 21123. 129. Hartel C, Osthues I, Rupp J, et al. Characterisation of the host inflammatory response to Staphylococcus epidermidis in neonatal whole blood. Arch Dis Child Fetal Neonatal Ed 2008;93(2):F1405. 130. Sikora JP, Chlebna-Sokol D, Krzyzanska-Oberbek A. Proinflammatory cytokines (IL-6, IL-8), cytokine inhibitors (IL-6sR, sTNFRII) and anti-inflammatory cytokines (IL-10, IL-13) in the pathogenesis of sepsis in newborns and infants. Arch Immunol Ther Exp (Warsz) 2001;49(5):399404. 131. Opal SM, Esmon CT. Bench-to-bedside review: functional relationships between coagulation and the innate immune response and their respective roles in the pathogenesis of sepsis. Crit Care 2003;7(1):2338. 132. Wang P, Wu P, Siegel MI, et al. Interleukin (IL)-10 inhibits nuclear factor kappa B (NF kappa B) activation in human monocytes. IL-10 and IL-4 suppress cytokine synthesis by different mechanisms. J Biol Chem 1995;270(16):955863. 133. Brubaker JO, Montaner LJ. Role of interleukin-13 in innate and adaptive immunity. Cell Mol Biol (Noisy-le-grand) 2001;47(4):63751. 134. Trepicchio WL, Bozza M, Pedneault G, et al. Recombinant human IL-11 attenuates the inflammatory response through down-regulation of proinflammatory cytokine release and nitric oxide production. J Immunol 1996;157(8):362734.

470

Wynn & Wong

135. Koj A. Termination of acute-phase response: role of some cytokines and antiinflammatory drugs. Gen Pharmacol 1998;31(1):918. 136. Spear ML, Stefano JL, Fawcett P, et al. Soluble interleukin-2 receptor as a predictor of neonatal sepsis. J Pediatr 1995;126(6):9825. 137. Dollner H, Vatten L, Linnebo I, et al. Inflammatory mediators in umbilical plasma from neonates who develop early-onset sepsis. Biol Neonate 2001; 80(1):417. 138. Buhimschi CS, Bhandari V, Han YW, et al. Using proteomics in perinatal and neonatal sepsis: hopes and challenges for the future. Curr Opin Infect Dis 2009;22(3):23543. 139. Bopp C, Hofer S, Weitz J, et al. sRAGE is elevated in septic patients and associated with patients outcome. J Surg Res 2008;147(1):7983. 140. Liliensiek B, Weigand MA, Bierhaus A, et al. Receptor for advanced glycation end products (RAGE) regulates sepsis but not the adaptive immune response. J Clin Invest 2004;113(11):164150. 141. Wolach B, Dolfin T, Regev R, et al. The development of the complement system after 28 weeks gestation. Acta Paediatr 1997;86(5):5237. 142. Notarangelo LD, Chirico G, Chiara A, et al. Activity of classical and alternative pathways of complement in preterm and small for gestational age infants. Pediatr Res 1984;18(3):2815. 143. Drossou V, Kanakoudi F, Diamanti E, et al. Concentrations of main serum opsonins in early infancy. Arch Dis Child Fetal Neonatal Ed 1995;72(3):F1725. 144. Miller ME, Stiehm ER. Phagocytic, opsonic and immunoglobulin studies in newborns. Calif Med 1973;119(2):4363. 145. Nupponen I, Andersson S, Jarvenpaa AL, et al. Neutrophil CD11b expression and circulating interleukin-8 as diagnostic markers for early-onset neonatal sepsis. Pediatrics 2001;108(1):E12. 146. Berger M, OShea J, Cross AS, et al. Human neutrophils increase expression of C3bi as well as C3b receptors upon activation. J Clin Invest 1984;74(5):156671. 147. Snyderman R, Goetzl EJ. Molecular and cellular mechanisms of leukocyte chemotaxis. Science 1981;213(4510):8307. 148. Vogt W. Anaphylatoxins: possible roles in disease. Complement 1986;3(3): 17788. 149. Ehlers MR. CR3: a general purpose adhesion-recognition receptor essential for innate immunity. Microbes Infect 2000;2(3):28994. 150. Markiewski MM, Lambris JD. The role of complement in inflammatory diseases from behind the scenes into the spotlight. Am J Pathol 2007;171(3):71527. 151. Nybo M, Sorensen O, Leslie R, et al. Reduced expression of C5a receptors on neutrophils from cord blood. Arch Dis Child Fetal Neonatal Ed 1998;78(2): F12932. 152. Lothian C, Dahlgren C, Lagercrantz H, et al. Different expression and mobilisation of the complement regulatory proteins CD35, CD55 and CD59 in neonatal and adult neutrophils. Biol Neonate 1997;72(1):1521. 153. Castellheim A, Lindenskov PH, Pharo A, et al. Meconium aspiration syndrome induces complement-associated systemic inflammatory response in newborn piglets. Scand J Immunol 2005;61(3):21725. 154. Wong HR, Doughty LA, Wedel N, et al. Plasma bactericidal/permeabilityincreasing protein concentrations in critically ill children with the sepsis syndrome. Pediatr Infect Dis J 1995;14(12):108791. 155. Levy O. Innate immunity of the newborn: basic mechanisms and clinical correlates. Nat Rev Immunol 2007;7(5):37990.

Treatment of Septic Shock in Neonates

471

156. Levy O, Martin S, Eichenwald E, et al. Impaired innate immunity in the newborn: newborn neutrophils are deficient in bactericidal/permeability-increasing protein. Pediatrics 1999;104(6):132733. 157. Michalek J, Svetlikova P, Fedora M, et al. Bactericidal permeability increasing protein gene variants in children with sepsis. Intensive Care Med 2007;33(12): 215864. 158. Dyke MP, Forsyth KD. Decreased plasma fibronectin concentrations in preterm infants with septicaemia. Arch Dis Child 1993;68(5 Spec No):55760. 159. Romeo MG, Tina LG, Sciacca A, et al. [Decreased plasma fibronectin (pFN) level in preterm infants with infections]. Pediatr Med Chir 1995;17(6):5636 [in Italian]. 160. Kalayci AG, Adam B, Yilmazer F, et al. The value of immunoglobulin and complement levels in the early diagnosis of neonatal sepsis. Acta Paediatr 1997;86(9):9991002. 161. Madden NP, Levinsky RJ, Bayston R, et al. Surgery, sepsis, and nonspecific immune function in neonates. J Pediatr Surg 1989;24(6):5626. 162. Hathaway WE, Mull MM, Pechet GS. Disseminated intravascular coagulation in the newborn. Pediatrics 1969;43(2):23340. 163. Kimberlin DW, Lin CY, Jacobs RF, et al. Natural history of neonatal herpes simplex virus infections in the acyclovir era. Pediatrics 2001;108(2):2239. 164. Rivers RP, Cattermole HE, Wright I. The expression of surface tissue factor apoprotein by blood monocytes in the course of infections in early infancy. Pediatr Res 1992;31(6):56773. 165. Markiewski MM, Nilsson B, Ekdahl KN, et al. Complement and coagulation: strangers or partners in crime? Trends Immunol 2007;28(4):18492. 166. Aronis S, Platokouki H, Photopoulos S, et al. Indications of coagulation and/or fibrinolytic system activation in healthy and sick very-low-birth-weight neonates. Biol Neonate 1998;74(5):33744. 167. Lauterbach R, Pawlik D, Radziszewska R, et al. Plasma antithrombin III and protein C levels in early recognition of late-onset sepsis in newborns. Eur J Pediatr 2006;165(9):5859. 168. Roman J, Velasco F, Fernandez F, et al. Coagulation, fibrinolytic and kallikrein systems in neonates with uncomplicated sepsis and septic shock. Haemostasis 1993;23(3):1428. 169. Venkataseshan S, Dutta S, Ahluwalia J, et al. Low plasma protein C values predict mortality in low birth weight neonates with septicemia. Pediatr Infect Dis J 2007;26(8):6848. 170. Sola MC, Del Vecchio A, Rimsza LM. Evaluation and treatment of thrombocytopenia in the neonatal intensive care unit. Clin Perinatol 2000;27(3):65579. 171. Guida JD, Kunig AM, Leef KH, et al. Platelet count and sepsis in very low birth weight neonates: is there an organism-specific response? Pediatrics 2003; 111(6 Pt 1):14115. 172. Finkelstein Y, Shenkman B, Sirota L, et al. Whole blood platelet deposition on extracellular matrix under flow conditions in preterm neonatal sepsis. Eur J Pediatr 2002;161(5):2704. 173. Bednarek FJ, Bean S, Barnard MR, et al. The platelet hyporeactivity of extremely low birth weight neonates is age-dependent. Thromb Res 2009;124(1):425. 174. Urlichs F, Speer C. Neutrophil function in preterm and term infants. Neoreviews 2004;5:e41730. 175. Marodi L. Innate cellular immune responses in newborns. Clin Immunol 2006; 118(23):13744.

472

Wynn & Wong

176. Christensen RD, Rothstein G. Exhaustion of mature marrow neutrophils in neonates with sepsis. J Pediatr 1980;96(2):3168. 177. Engle WA, McGuire WA, Schreiner RL, et al. Neutrophil storage pool depletion in neonates with sepsis and neutropenia. J Pediatr 1988;113(4):7479. 178. Sarkar S, Bhagat I, Hieber S, et al. Can neutrophil responses in very low birth weight infants predict the organisms responsible for late-onset bacterial or fungal sepsis? J Perinatol 2006;26(8):5015. 179. Christensen RD, Bradley PP, Rothstein G. The leukocyte left shift in clinical and experimental neonatal sepsis. J Pediatr 1981;98(1):1015. 180. Drossou V, Kanakoudi F, Tzimouli V, et al. Impact of prematurity, stress and sepsis on the neutrophil respiratory burst activity of neonates. Biol Neonate 1997;72(4):2019. 181. Shigeoka AO, Santos JI, Hill HR. Functional analysis of neutrophil granulocytes from healthy, infected, and stressed neonates. J Pediatr 1979;95(3): 45460. 182. Wright WC Jr, Ank BJ, Herbert J, et al. Decreased bactericidal activity of leukocytes of stressed newborn infants. Pediatrics 1975;56(4):57984. 183. Linderkamp O, Ruef P, Brenner B, et al. Passive deformability of mature, immature, and active neutrophils in healthy and septicemic neonates. Pediatr Res 1998;44(6):94650. 184. Mease AD, Burgess DP, Thomas PJ. Irreversible neutrophil aggregation. A mechanism of decreased newborn neutrophil chemotactic response. Am J Pathol 1981;104(1):98102. 185. Ohman L, Tullus K, Katouli M, et al. Correlation between susceptibility of infants to infections and interaction with neutrophils of Escherichia coli strains causing neonatal and infantile septicemia. J Infect Dis 1995;171(1):12833. 186. Allgaier B, Shi M, Luo D, et al. Spontaneous and Fas-mediated apoptosis are diminished in umbilical cord blood neutrophils compared with adult neutrophils. J Leukoc Biol 1998;64(3):3316. 187. Hanna N, Vasquez P, Pham P, et al. Mechanisms underlying reduced apoptosis in neonatal neutrophils. Pediatr Res 2005;57(1):5662. 188. Koenig JM, Stegner JJ, Schmeck AC, et al. Neonatal neutrophils with prolonged survival exhibit enhanced inflammatory and cytotoxic responsiveness. Pediatr Res 2005;57(3):4249. 189. Nupponen I, Pesonen E, Andersson S, et al. Neutrophil activation in preterm infants who have respiratory distress syndrome. Pediatrics 2002;110(1 Pt 1):3641. 190. Nathan CF. Secretory products of macrophages. J Clin Invest 1987;79(2): 31926. 191. Hodge G, Hodge S, Han P, et al. Multiple leucocyte activation markers to detect neonatal infection. Clin Exp Immunol 2004;135(1):1259. 192. Georgeson GD, Szony BJ, Streitman K, et al. Natural killer cell cytotoxicity is deficient in newborns with sepsis and recurrent infections. Eur J Pediatr 2001; 160(8):47882. 193. el-Sameea ER, Metwally SS, Mashhour E, et al. Evaluation of natural killer cells as diagnostic markers of early onset neonatal sepsis: comparison with C-reactive protein and interleukin-8. Egypt J Immunol 2004;11(1):91102. 194. Mazur B, Godula-Stuglik U, Domarecki A, et al. [The influence of severe infections on the natural killer cells in neonates with vary gestational age]. Ginekol Pol 2000;71(6):5429 [in Polish]. 195. Marshall JS, Jawdat DM. Mast cells in innate immunity. J Allergy Clin Immunol 2004;114(1):217.

Treatment of Septic Shock in Neonates

473

196. von Kockritz-Blickwede M, Goldmann O, Thulin P, et al. Phagocytosis-independent antimicrobial activity of mast cells by means of extracellular trap formation. Blood 2008;111(6):307080. 197. Mazzoni A, Young HA, Spitzer JH, et al. Histamine regulates cytokine production in maturing dendritic cells, resulting in altered T cell polarization. J Clin Invest 2001;108(12):186573. 198. Damsgaard TE, Nielsen BW, Henriques U, et al. Histamine releasing cells of the newborn. Mast cells from the umbilical cord matrix and basophils from cord blood. Pediatr Allergy Immunol 1996;7(2):8390. 199. Schneider LA, Schlenner SM, Feyerabend TB, et al. Molecular mechanism of mast cell mediated innate defense against endothelin and snake venom sarafotoxin. J Exp Med 2007;204(11):262939. 200. Peters K, Unger RE, Brunner J, et al. Molecular basis of endothelial dysfunction in sepsis. Cardiovasc Res 2003;60(1):4957. 201. Marshall JC. Such stuff as dreams are made on: mediator-directed therapy in sepsis. Nat Rev Drug Discov 2003;2(5):391405. 202. Marom D, Yuhas Y, Sirota L, et al. Nitric oxide levels in preterm and term infants and in premature infants with bacteremia. Biol Neonate 2004;86(3):1604. 203. Schrama AJ, de Beaufort AJ, Poorthuis BJ, et al. Secretory phospholipase A(2) in newborn infants with sepsis. J Perinatol 2008;28(4):2916. 204. Siauw C, Kobsar A, Dornieden C, et al. Group B streptococcus isolates from septic patients and healthy carriers differentially activate platelet signaling cascades. Thromb Haemost 2006;95(5):83649. 205. Figueras-Aloy J, Gomez-Lopez L, Rodriguez-Miguelez JM, et al. Plasma endothelin-1 and clinical manifestations of neonatal sepsis. J Perinat Med 2004;32(6): 5226. 206. Kuhl PG, Cotton RB, Schweer H, et al. Endogenous formation of prostanoids in neonates with persistent pulmonary hypertension. Arch Dis Child 1989;64(7 Spec No):94952. 207. Rosenberg AA, Kennaugh J, Koppenhafer SL, et al. Elevated immunoreactive endothelin-1 levels in newborn infants with persistent pulmonary hypertension. J Pediatr 1993;123(1):10914. 208. Rossi P, Persson B, Boels PJ, et al. Endotoxemic pulmonary hypertension is largely mediated by endothelin-induced venous constriction. Intensive Care Med 2008;34(5):87380. 209. Bulger EM, Maier RV. Lipid mediators in the pathophysiology of critical illness. Crit Care Med 2000;28(Suppl 4):N2736. 210. Gotsch F, Romero R, Kusanovic JP, et al. The fetal inflammatory response syndrome. Clin Obstet Gynecol 2007;50(3):65283. 211. Shi Y, Li HQ, Shen CK, et al. Plasma nitric oxide levels in newborn infants with sepsis. J Pediatr 1993;123(3):4358. 212. Kultursay N, Kantar M, Akisu M, et al. Platelet-activating factor concentrations in healthy and septic neonates. Eur J Pediatr 1999;158(9):7401. 213. Lopez A, Lorente JA, Steingrub J, et al. Multiple-center, randomized, placebocontrolled, double-blind study of the nitric oxide synthase inhibitor 546C88: effect on survival in patients with septic shock. Crit Care Med 2004;32(1):2130. 214. Nimah M, Brilli RJ. Coagulation dysfunction in sepsis and multiple organ system failure. Crit Care Clin 2003;19(3):44158. 215. Cribbs SK, Martin GS, Rojas M. Monitoring of endothelial dysfunction in critically ill patients: the role of endothelial progenitor cells. Curr Opin Crit Care 2008; 14(3):35460.

474

Wynn & Wong

216. Gibson RL, Nizet V, Rubens CE. Group B streptococcal beta-hemolysin promotes injury of lung microvascular endothelial cells. Pediatr Res 1999;45(5 Pt 1):62634. 217. Dakshinamurti S. Pathophysiologic mechanisms of persistent pulmonary hypertension of the newborn. Pediatr Pulmonol 2005;39(6):492503. 218. Arai H, Matsuda T, Goto R, et al. Increased numbers of macrophages in tracheal aspirates in premature infants with funisitis. Pediatr Int 2008;50(2):1848. 219. McKiernan CA, Lieberman SA. Circulatory shock in children: an overview. Pediatr Rev 2005;26(12):45160. 220. Luce WA, Hoffman TM, Bauer JA. Bench-to-bedside review: developmental influences on the mechanisms, treatment and outcomes of cardiovascular dysfunction in neonatal versus adult sepsis. Crit Care 2007;11(5):228. 221. Maeder M, Fehr T, Rickli H, et al. Sepsis-associated myocardial dysfunction: diagnostic and prognostic impact of cardiac troponins and natriuretic peptides. Chest 2006;129(5):134966. 222. Tabbutt S. Heart failure in pediatric septic shock: utilizing inotropic support. Crit Care Med 2001;29(Suppl 10):S2316. 223. Ceneviva G, Paschall JA, Maffei F, et al. Hemodynamic support in fluid-refractory pediatric septic shock. Pediatrics 1998;102(2):e19. 224. Carcillo JA, Fields AI. Clinical practice parameters for hemodynamic support of pediatric and neonatal patients in septic shock. Crit Care Med 2002;30(6):136578. 225. Evans N. Which inotrope for which baby? Arch Dis Child Fetal Neonatal Ed 2006;91(3):F21320. 226. Kluckow M, Evans N. Relationship between blood pressure and cardiac output in preterm infants requiring mechanical ventilation. J Pediatr 1996;129(4):50612. 227. Seri I, Noori S. Diagnosis and treatment of neonatal hypotension outside the transitional period. Early Hum Dev 2005;81(5):40511. 228. Cochat P, Bourgeois J, Gilly J, et al. [Anatomical study of the kidneys of newborn infants dying after a septic state]. Pediatrie 1986;41(1):715 [in French]. 229. Csaicsich D, Russo-Schlaff N, Messerschmidt A, et al. Renal failure, comorbidity and mortality in preterm infants. Wien Klin Wochenschr 2008;120(56):1537. 230. Spapen H. Liver perfusion in sepsis, septic shock, and multiorgan failure. Anat Rec (Hoboken) 2008;291(6):71420. 231. Bhutta ZA, Yusuf K. Neonatal sepsis in Karachi: factors determining outcome and mortality. J Trop Pediatr 1997;43(2):6570. 232. Faix RG, Donn SM. Association of septic shock caused by early-onset group B streptococcal sepsis and periventricular leukomalacia in the preterm infant. Pediatrics 1985;76(3):4159. 233. Harris MC, DAngio CT, Gallagher PR, et al. Cytokine elaboration in critically ill infants with bacterial sepsis, necrotizing entercolitis, or sepsis syndrome: correlation with clinical parameters of inflammation and mortality. J Pediatr 2005; 147(4):4628. 234. Di Naro E, Cromi A, Ghezzi F, et al. Fetal thymic involution: a sonographic marker of the fetal inflammatory response syndrome. Am J Obstet Gynecol 2006;194(1):1539. 235. Cinel I, Dellinger RP. Advances in pathogenesis and management of sepsis. Curr Opin Infect Dis 2007;20(4):34552. 236. Cinel I, Opal SM. Molecular biology of inflammation and sepsis: a primer. Crit Care Med 2009;37(1):291304. 237. Pfenninger J, Tschaeppeler H, Wagner BP, et al. The paradox of adult respiratory distress syndrome in neonates. Pediatr Pulmonol 1991;10(1):1824.

Treatment of Septic Shock in Neonates

475

238. Engle WA. Surfactant-replacement therapy for respiratory distress in the preterm and term neonate. Pediatrics 2008;121(2):41932. 239. Ng PC, Lam CW, Fok TF, et al. Refractory hypotension in preterm infants with adrenocortical insufficiency. Arch Dis Child Fetal Neonatal Ed 2001;84(2):F1224. 240. Das BK, Agarwal P, Agarwal JK, et al. Serum cortisol and thyroid hormone levels in neonates with sepsis. Indian J Pediatr 2002;69(8):6635. 241. Glavina-Durdov M, Springer O, Capkun V, et al. The grade of acute thymus involution in neonates correlates with the duration of acute illness and with the percentage of lymphocytes in peripheral blood smear. Pathological study. Biol Neonate 2003;83(4):22934. 242. Itoh K, Aihara H, Takada S, et al. Clinicopathological differences between earlyonset and late-onset sepsis and pneumonia in very low birth weight infants. Pediatr Pathol 1990;10(5):75768. 243. Timens W, Boes A, Rozeboom-Uiterwijk T, et al. Immaturity of the human splenic marginal zone in infancy. Possible contribution to the deficient infant immune response. J Immunol 1989;143(10):32006. 244. Toti P, De Felice C, Occhini R, et al. Spleen depletion in neonatal sepsis and chorioamnionitis. Am J Clin Pathol 2004;122(5):76571. 245. Toti P, De Felice C, Stumpo M, et al. Acute thymic involution in fetuses and neonates with chorioamnionitis. Hum Pathol 2000;31(9):11218. 246. van Baarlen J, Schuurman HJ, Huber J. Acute thymus involution in infancy and childhood: a reliable marker for duration of acute illness. Hum Pathol 1988; 19(10):115560. 247. Hotchkiss RS, Coopersmith CM, Karl IE. Prevention of lymphocyte apoptosis a potential treatment of sepsis? Clin Infect Dis 2005;41(Suppl 7):S4659. 248. Kalman L, Lindegren ML, Kobrynski L, et al. Mutations in genes required for T-cell development: IL7R, CD45, IL2RG, JAK3, RAG1, RAG2, ARTEMIS, and ADA and severe combined immunodeficiency: HuGE review. Genet Med 2004;6(1):1626. 249. Le Tulzo Y, Pangault C, Gacouin A, et al. Early circulating lymphocyte apoptosis in human septic shock is associated with poor outcome. Shock 2002;18(6): 48794. 250. Wynn JL, Scumpia PO, Winfield RD, et al. Defective innate immunity predisposes murine neonates to poor sepsis outcome but is reversed by TLR agonists. Blood 2008;112(5):17508. 251. Bauer J, Hentschel R, Linderkamp O. Effect of sepsis syndrome on neonatal oxygen consumption and energy expenditure. Pediatrics 2002;110(6):e69. 252. Romeo C, Eaton S, Spitz L, et al. Nitric oxide inhibits neonatal hepatocyte oxidative metabolism. J Pediatr Surg 2000;35(1):448. 253. Eaton S. Impaired energy metabolism during neonatal sepsis: the effects of glutamine. Proc Nutr Soc 2003;62(3):74551. 254. Dellinger RP, Levy MM, Carlet JM, et al. Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock: 2008. Intensive Care Med 2008;34(1):1760. 255. Brierley J, Carcillo JA, Choong K, et al. Clinical practice parameters for hemodynamic support of pediatric and neonatal septic shock: 2007 update from the American College of Critical Care Medicine. Crit Care Med 2009;37(2):66688. 256. Parker MM, Hazelzet JA, Carcillo JA. Pediatric considerations. Crit Care Med 2004;32(Suppl 11):S5914. 257. Dempsey EM, Al Hazzani F, Barrington KJ. Permissive hypotension in the extremely low birthweight infant with signs of good perfusion. Arch Dis Child Fetal Neonatal Ed 2009;94(4):F2414.

476

Wynn & Wong

258. Shiao SY. Effects of fetal hemoglobin on accurate measurements of oxygen saturation in neonates. J Perinat Neonatal Nurs 2005;19(4):34861. 259. Kluckow M, Seri I, Evans N. Echocardiography and the neonatologist. Pediatr Cardiol 2008;29(6):10437. 260. Kluckow M, Seri I, Evans N. Functional echocardiography: an emerging clinical tool for the neonatologist. J Pediatr 2007;150(2):12530. 261. Evans N, Kluckow M, Simmons M, et al. Which to measure, systemic or organ blood flow? Middle cerebral artery and superior vena cava flow in very preterm infants. Arch Dis Child Fetal Neonatal Ed 2002;87(3):F1814. 262. Kluckow M, Evans N. Superior vena cava flow in newborn infants: a novel marker of systemic blood flow. Arch Dis Child Fetal Neonatal Ed 2000;82(3):F1827. 263. Hunt RW, Evans N, Rieger I, et al. Low superior vena cava flow and neurodevelopment at 3 years in very preterm infants. J Pediatr 2004;145(5):58892. 264. Miletin J, Pichova K, Dempsey EM. Bedside detection of low systemic flow in the very low birth weight infant on day 1 of life. Eur J Pediatr 2009;168(7): 80913. 265. Fortune PM, Wagstaff M, Petros AJ. Cerebro-splanchnic oxygenation ratio (CSOR) using near infrared spectroscopy may be able to predict splanchnic ischaemia in neonates. Intensive Care Med 2001;27(8):14017. 266. Johnson BA, Hoffman GM, Tweddell JS, et al. Near-infrared spectroscopy in neonates before palliation of hypoplastic left heart syndrome. Ann Thorac Surg 2009;87(2):5717 [discussion: 5779]. 267. Osborn DA, Evans N, Kluckow M, et al. Low superior vena cava flow and effect of inotropes on neurodevelopment to 3 years in preterm infants. Pediatrics 2007; 120(2):37280. 268. Seri I. Circulatory support of the sick preterm infant. Semin Neonatol 2001;6(1): 8595. 269. Munro MJ, Walker AM, Barfield CP. Hypotensive extremely low birth weight infants have reduced cerebral blood flow. Pediatrics 2004;114(6):15916. 270. Han YY, Carcillo JA, Dragotta MA, et al. Early reversal of pediatric-neonatal septic shock by community physicians is associated with improved outcome. Pediatrics 2003;112(4):7939. 271. Osborn DA, Evans N. Early volume expansion for prevention of morbidity and mortality in very preterm infants [review]. Cochrane Database Syst Rev 2004; 2:CD002055. 272. Goldberg RN, Chung D, Goldman SL, et al. The association of rapid volume expansion and intraventricular hemorrhage in the preterm infant. J Pediatr 1980;96(6):10603. 273. Pellicer A, Valverde E, Elorza MD, et al. Cardiovascular support for low birth weight infants and cerebral hemodynamics: a randomized, blinded, clinical trial. Pediatrics 2005;115(6):150112. 274. Osborn D, Evans N, Kluckow M. Randomized trial of dobutamine versus dopamine in preterm infants with low systemic blood flow. J Pediatr 2002;140(2): 18391. 275. Osborn DA, Paradisis M, Evans N. The effect of inotropes on morbidity and mortality in preterm infants with low systemic or organ blood flow [review]. Cochrane Database Syst Rev 2007;1:CD005090. 276. Barton P, Garcia J, Kouatli A, et al. Hemodynamic effects of i.v. milrinone lactate in pediatric patients with septic shock. A prospective, double-blinded, randomized, placebo-controlled, interventional study. Chest 1996;109(5): 130212.

Treatment of Septic Shock in Neonates

477

277. Paradisis M, Evans N, Kluckow M, et al. Randomized trial of milrinone versus placebo for prevention of low systemic blood flow in very preterm infants. J Pediatr 2009;154(2):18995. 278. Leone M, Martin C. Role of terlipressin in the treatment of infants and neonates with catecholamine-resistant septic shock. Best Pract Res Clin Anaesthesiol 2008;22(2):32333. 279. Meyer S, Gottschling S, Baghai A, et al. Arginine-vasopressin in catecholaminerefractory septic versus non-septic shock in extremely low birth weight infants with acute renal injury. Crit Care 2006;10(3):R71. 280. Annane D, Bellissant E, Bollaert PE, et al. Corticosteroids for severe sepsis and septic shock: a systematic review and meta-analysis. BMJ 2004; 329(7464):480. 281. Annane D, Sebille V, Charpentier C, et al. Effect of treatment with low doses of hydrocortisone and fludrocortisone on mortality in patients with septic shock. JAMA 2002;288(7):86271. 282. Sprung CL, Annane D, Keh D, et al. Hydrocortisone therapy for patients with septic shock. N Engl J Med 2008;358(2):11124. 283. Togari H, Sugiyama S, Ogino T, et al. Interactions of endotoxin with cortisol and acute phase proteins in septic shock neonates. Acta Paediatr Scand 1986; 75(1):6974. 284. Seri I, Tan R, Evans J. Cardiovascular effects of hydrocortisone in preterm infants with pressor-resistant hypotension. Pediatrics 2001;107(5):10704. 285. Noori S, Friedlich P, Wong P, et al. Hemodynamic changes after low-dosage hydrocortisone administration in vasopressor-treated preterm and term neonates. Pediatrics 2006;118(4):145666. 286. Fernandez EF, Watterberg KL. Relative adrenal insufficiency in the preterm and term infant. J Perinatol 2009;29(Suppl 2):S449. 287. Roberts JD Jr, Fineman JR, Morin FC 3rd, et al. Inhaled nitric oxide and persistent pulmonary hypertension of the newborn. The Inhaled Nitric Oxide Study Group. N Engl J Med 1997;336(9):60510. 288. Farrow KN, Fliman P, Steinhorn RH. The diseases treated with ECMO: focus on PPHN. Semin Perinatol 2005;29(1):814. 289. Maclaren G, Butt W. Extracorporeal membrane oxygenation and sepsis. Crit Care Resusc 2007;9(1):7680. 290. Shankaran S, Laptook AR, Ehrenkranz RA, et al. Whole-body hypothermia for neonates with hypoxic-ischemic encephalopathy. N Engl J Med 2005;353(15): 157484. 291. Emery EF, Greenough A, Gamsu HR. Randomised controlled trial of colloid infusions in hypotensive preterm infants. Arch Dis Child 1992;67(10 Spec No): 11858. 292. Neu J, Huang Y. Nutrition of premature and critically ill neonates. Nestle Nutr Workshop Ser Clin Perform Programme 2003;8:17181 [discussion: 1815]. 293. Park W, Paust H, Schroder H. Lipid infusion in premature infants suffering from sepsis. JPEN J Parenter Enteral Nutr 1984;8(3):2902. 294. Toce SS, Keenan WJ. Lipid intolerance in newborns is associated with hepatic dysfunction but not infection. Arch Pediatr Adolesc Med 1995;149(11):124953. 295. Basu R, Muller DP, Eaton S, et al. Lipid peroxidation can be reduced in infants on total parenteral nutrition by promoting fat utilisation. J Pediatr Surg 1999;34(2): 2559. 296. Pierro A. Metabolism and nutritional support in the surgical neonate. J Pediatr Surg 2002;37(6):81122.

478

Wynn & Wong

297. Mathur NB, Singh A, Sharma VK, et al. Evaluation of risk factors for fatal neonatal sepsis. Indian Pediatr 1996;33(10):81722. 298. Adams-Chapman I, Stoll BJ. Neonatal infection and long-term neurodevelopmental outcome in the preterm infant. Curr Opin Infect Dis 2006;19(3): 2907. 299. Stoll BJ, Hansen NI, Adams-Chapman I, et al. Neurodevelopmental and growth impairment among extremely low-birth-weight infants with neonatal infection. JAMA 2004;292(19):235765. 300. Bassler D, Stoll BJ, Schmidt B, et al. Using a count of neonatal morbidities to predict poor outcome in extremely low birth weight infants: added role of neonatal infection. Pediatrics 2009;123(1):3138. 301. Shah DK, Doyle LW, Anderson PJ, et al. Adverse neurodevelopment in preterm infants with postnatal sepsis or necrotizing enterocolitis is mediated by white matter abnormalities on magnetic resonance imaging at term. J Pediatr 2008; 153(2):1705, 175 e171. 302. Hintz SR, Kendrick DE, Stoll BJ, et al. Neurodevelopmental and growth outcomes of extremely low birth weight infants after necrotizing enterocolitis. Pediatrics 2005;115(3):696703. 303. Manzoni P, Maestri A, Leonessa M, et al. Fungal and bacterial sepsis and threshold ROP in preterm very low birth weight neonates. J Perinatol 2006; 26(1):2330. 304. Wong HR, Cvijanovich N, Lin R, et al. Identification of pediatric septic shock subclasses based on genome-wide expression profiling. BMC Med 2009;7:34. 305. Wong HR, Cvijanovich N, Allen GL, et al. Genomic expression profiling across the pediatric systemic inflammatory response syndrome, sepsis, and septic shock spectrum. Crit Care Med 2009;37(5):155866. 306. Wong HR, Odoms K, Sakthivel B. Divergence of canonical danger signals: the genome-level expression patterns of human mononuclear cells subjected to heat shock or lipopolysaccharide. BMC Immunol 2008;9:24. 307. Cvijanovich N, Shanley TP, Lin R, et al. Validating the genomic signature of pediatric septic shock. Physiol Genomics 2008;34(1):12734. 308. Shanley TP, Cvijanovich N, Lin R, et al. Genome-level longitudinal expression of signaling pathways and gene networks in pediatric septic shock. Mol Med 2007; 13(910):495508. 309. Tang BM, McLean AS, Dawes IW, et al. Gene-expression profiling of Gram-positive and Gram-negative sepsis in critically ill patients. Crit Care Med 2008;36(4): 11258. 310. Tang BM, McLean AS, Dawes IW, et al. The use of gene-expression profiling to identify candidate genes in human sepsis. Am J Respir Crit Care Med 2007; 176(7):67684. 311. Tang BM, McLean AS, Dawes IW, et al. Gene-expression profiling of peripheral blood mononuclear cells in sepsis. Crit Care Med 2009;37(3):8828. 312. Karvunidis T, Mares J, Thongboonkerd V, et al. Recent progress of proteomics in critical illness. Shock 2009;31(6):54552. 313. Buhimschi CS, Bhandari V, Hamar BD, et al. Proteomic profiling of the amniotic fluid to detect inflammation, infection, and neonatal sepsis. PLoS Med 2007;4(1):e18. 314. Feezor RJ, Cheng A, Paddock HN, et al. Functional genomics and gene expression profiling in sepsis: beyond class prediction. Clin Infect Dis 2005; 41(Suppl 7):S42735. 315. Levy O, Sisson RB, Kenyon J, et al. Enhancement of neonatal innate defense: effects of adding an N-terminal recombinant fragment of bactericidal/

Treatment of Septic Shock in Neonates

479

316.

317.

318. 319. 320.

321.

322. 323.

permeability-increasing protein on growth and tumor necrosis factor-inducing activity of Gram-negative bacteria tested in neonatal cord blood ex vivo. Infect Immun 2000;68(9):51205. Reinhart K, Gluck T, Ligtenberg J, et al. CD14 receptor occupancy in severe sepsis: results of a phase I clinical trial with a recombinant chimeric CD14 monoclonal antibody (IC14). Crit Care Med 2004;32(5):11008. Lauterbach R, Pawlik D, Kowalczyk D, et al. Effect of the immunomodulating agent, pentoxifylline, in the treatment of sepsis in prematurely delivered infants: a placebo-controlled, double-blind trial. Crit Care Med 1999;27(4):80714. Tracey KJ. Physiology and immunology of the cholinergic antiinflammatory pathway. J Clin Invest 2007;117(2):28996. Pleiner J, Schaller G, Mittermayer F, et al. Simvastatin prevents vascular hyporeactivity during inflammation. Circulation 2004;110(21):334954. Ikegami M, Carter K, Bishop K, et al. Intratracheal recombinant surfactant protein d prevents endotoxin shock in the newborn preterm lamb. Am J Respir Crit Care Med 2006;173(12):13427. Mohan P, Abrams SA. Oral lactoferrin for the treatment of sepsis and necrotizing enterocolitis in neonates [review]. Cochrane Database Syst Rev 2009;1: CD007138. Venkatesh M, Abrams S. Can lactoferrin prevent neonatal sepsis and necrotizing enterocolitis? Expert Rev Anti Infect Ther 2009;7(5):51525. Suri M, Harrison L, Van de Ven C, et al. Immunotherapy in the prophylaxis and treatment of neonatal sepsis. Curr Opin Pediatr 2003;15(2):15560.

Você também pode gostar